CN114010775A - Group B streptococcal polysaccharide-protein conjugates, methods of making conjugates, immunogenic compositions containing conjugates, and uses thereof - Google Patents

Group B streptococcal polysaccharide-protein conjugates, methods of making conjugates, immunogenic compositions containing conjugates, and uses thereof Download PDF

Info

Publication number
CN114010775A
CN114010775A CN202111336096.5A CN202111336096A CN114010775A CN 114010775 A CN114010775 A CN 114010775A CN 202111336096 A CN202111336096 A CN 202111336096A CN 114010775 A CN114010775 A CN 114010775A
Authority
CN
China
Prior art keywords
polysaccharide
gbs
conjugate
serotype
capsular polysaccharide
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202111336096.5A
Other languages
Chinese (zh)
Inventor
A·S·安德森
巴拉 A·S·布彭德尔
R·G·K·唐纳德
顾建新
K·U·詹森
R·K·凯因坦
L·康德克
金进焕
P·利伯拉托尔
A·K·普拉萨德
M·E·鲁彭
I·L·斯卡利
S·辛格
C·X·杨
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Pfizer Inc
Original Assignee
Pfizer Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Pfizer Inc filed Critical Pfizer Inc
Publication of CN114010775A publication Critical patent/CN114010775A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/09Lactobacillales, e.g. aerococcus, enterococcus, lactobacillus, lactococcus, streptococcus
    • A61K39/092Streptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/385Haptens or antigens, bound to carriers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/40Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum bacterial
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/6415Toxins or lectins, e.g. clostridial toxins or Pseudomonas exotoxins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/646Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent the entire peptide or protein drug conjugate elicits an immune response, e.g. conjugate vaccines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • C07K16/1267Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria
    • C07K16/1275Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria from Gram-positive bacteria from Streptococcus (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/44Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material not provided for elsewhere, e.g. haptens, metals, DNA, RNA, amino acids
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B37/00Preparation of polysaccharides not provided for in groups C08B1/00 - C08B35/00; Derivatives thereof
    • C08B37/0003General processes for their isolation or fractionation, e.g. purification or extraction from biomass
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/55Medicinal preparations containing antigens or antibodies characterised by the host/recipient, e.g. newborn with maternal antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55505Inorganic adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55566Emulsions, e.g. Freund's adjuvant, MF59
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/555Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
    • A61K2039/55511Organic adjuvants
    • A61K2039/55577Saponins; Quil A; QS21; ISCOMS
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/60Medicinal preparations containing antigens or antibodies characteristics by the carrier linked to the antigen
    • A61K2039/6031Proteins
    • A61K2039/6037Bacterial toxins, e.g. diphteria toxoid [DT], tetanus toxoid [TT]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/70Multivalent vaccine
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Organic Chemistry (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biochemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Virology (AREA)
  • Toxicology (AREA)
  • Sustainable Development (AREA)
  • Materials Engineering (AREA)
  • Polymers & Plastics (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention relates to immunogenic polysaccharide-protein conjugates comprising a Capsular Polysaccharide (CP) from streptococcus agalactiae, commonly known as Group B Streptococcus (GBS), and a carrier protein, wherein the CP is selected from the group consisting of: serotypes Ia, Ib, II, III, IV, V, VI, VII, VIII and IX and wherein the sialic acid level of CP is greater than about 60%. The invention also relates to methods of making the conjugates and immunogenic compositions comprising the conjugates. The invention also relates to immunogenic compositions comprising a polysaccharide-protein conjugate, wherein the conjugate comprises a CP from GBS serotype IV and at least one additional serotype. The invention further relates to methods of inducing an immune response to GBS in a subject and/or reducing or preventing an aggressive GBS disease in a subject using the compositions disclosed herein. The antibodies produced can be used to treat or prevent GBS infection by passive immunotherapy.

Description

Group B streptococcal polysaccharide-protein conjugates, methods of making conjugates, immunogenic compositions containing conjugates, and uses thereof
The present application is a divisional application of chinese patent application 201680025709.4 entitled "group B streptococcus polysaccharide-protein conjugates, methods of making conjugates, immunogenic compositions containing conjugates and uses thereof" filed 2016, 29/4.
Technical Field
The present invention relates to immunogenic polysaccharide-protein conjugates comprising a Capsular Polysaccharide (CP) from Streptococcus agalactiae (generally known as Group B Streptococcus (GBS)) and a carrier protein, wherein the CP is selected from the group consisting of: serotypes Ia, Ib, II, III, IV, V, VI, VII, VIII and IX, and wherein the amount of sialic acid (sialic acid) of the CP is greater than about 60%. The invention also relates to methods of making the conjugates and immunogenic compositions comprising the conjugates. The invention also relates to immunogenic compositions comprising a polysaccharide-protein conjugate, wherein the conjugate comprises a CP from GBS serotype IV and at least one additional serotype. The invention further relates to methods of inducing an immune response to GBS in a subject and/or reducing or preventing an aggressive GBS disease in a subject using the compositions disclosed herein. The antibodies produced can be used to treat or prevent GBS infection by passive immunotherapy.
Background
Streptococcus agalactiae is a gram-positive polysaccharide-encapsulated organism, also known as Group B Streptococcus (GBS). They are common symbionts (commenal) of the human gastrointestinal and reproductive tracts and also responsible for serious diseases in infants and elderly (Baker, c.j., Vaccine,31(suppl.4): D3-D6 (2013)). The main risk factor for GBS infection in infants is maternal colonization (Dillon, h.c., et al, j.pediatr, 110(1):31-36 (1987)). As many as one quarter of women have GBS in their recto-vaginal tract, which can infect amniotic fluid or infants before or during parturition causing sepsis, pneumonia and meningitis (Baker 2013; Heath, p.t., et al, BMJ clin.evid. (Online), pii:0323 (2014)). 25% of infants surviving GBS meningitis suffer from neurological impairment, 19% of which experience cognitive delay (cognitive delay), cerebral palsy, blindness and hearing loss (Libster, r., et al, Pediatrics,130(1): e8-152012 (2012)). GBS also causes abortion and premature birth, and is associated with stillbirth (McDonald, H.M., et al, Infectious Diseases in Obstotris and Gynecology,8(5-6): 220-. The risk of infection is much higher in very low birth weight infants, with up to 3% infection and up to 30% mortality (even if treated immediately with antibiotics) (Heath 2014).
GBS screening and antibiotic prophylaxis (IAP) at production, introduced late in the 1990 s in the united states, demonstrated a reduction in the rate of neonatal disease (early onset disease [ EOD ]) occurring within the first week of birth, but had no measurable effect on the rate of Late Onset Disease (LOD) occurring later in the first 3 months of birth. The current ratio of EOD and LOD cases in the united states is 0.25 and 0.27 per 1000 births (Centers for Disease Control and Prevention) (CDC), respectively, viable bacteria Core (ABC) supervision Report (Active Bacterial Core survey Report) (2013), available at http:// www.cdc.gov/ABCs/reports-definitions/subversions/gs13. pdf). After introduction of pneumococcal conjugate vaccines for the prevention of invasive pneumococcal disease, including bacteremia and meningitis, although IAPs are useful in the prevention of GBS disease, GBS has become the most common single cause of neonatal sepsis (EOD) and meningitis (<2mo) in the united states (Verani, j.r., et al, MMWR,59(RR10):1-32 (2010); thigh, m.c., et al, New England Journal of Medicine,364(21): 2016-. Unlike in The United states, introduction of control guidelines and IAPs for aggressive GBS Diseases does not reduce The incidence of EOD in The Netherlands or British (Bekker, V., et al, The Lancet Infections Diseases,14(11): 1083-. This lack of effect may be due to lack of comprehensive screening and IAP is limited to mothers of the highest risk group (e.g. fever, delayed rupture of membranes). The proportion of EOD in countries where IAP is not used is significantly higher, with an average incidence reported of 0.75 (95% CI 0.58-0.89) per 1000 live births (Edmond, K.M, et al, Lancet,379(9815):547-556 (2012)).
Another population at risk for GBS disease is the elderly. Risk factors include chronic medical problems such as diabetes, cancer, heart failure, neurological and urological conditions. According to CDC ABC surveillance data, the annual incidence of invasive GBS in 2013 in the U.S. is 0.28 per 1,000 adults or 12,400 cases per year in adults ≧ 65. This ratio approximates the incidence of invasive pneumococcal disease in the elderly (0.30/1,000 versus > 65). These ratios are expected to continue to increase in the United states and Europe (CDC 2013; Lamagni 2013).
One method of preventing GBS disease in infants and the elderly is the use of polysaccharide based vaccines. In the united states, administration of maternal GBS prophylactic vaccines has the potential to prevent GBS disease in infants, whether or not IAPs are used. Although polysaccharides can be immunogenic themselves, conjugation of polysaccharides to protein carriers has been used to improve immunogenicity, particularly for infants and the elderly. Polysaccharide-protein conjugate vaccines are manufactured using polysaccharides (usually from the bacterial S-layer) linked to a protein carrier. Chemical bonding of polysaccharides to protein carriers can induce an immune response against bacteria whose surfaces exhibit the polysaccharides contained in the vaccine, thereby preventing disease. Thus, vaccination with polysaccharides from pathogenic bacteria is a possible strategy to boost host immunity.
The polysaccharide covering the bacteria varies greatly, even in a single species. For example, there are ten different serotypes of GBS due to variation in the bacterial polysaccharide capsule. Therefore, it is desirable that polysaccharide-based vaccines consist of a group of polysaccharides to ensure a broad coverage of different circulating strains.
The carrier protein may be a related protein antigen from the pathogen of interest, boosting a specific immune response to the pathogen, or may be a general immunogenic protein that also serves as an adjuvant or general immune response stimulator.
Individual monovalent polysaccharide-protein conjugates of GBS serotypes Ia, Ib, II, III and V have been evaluated in phase 1 and phase 2 clinical trials in non-pregnant adults (Brigtsen, A.K., et al, Journal of Infectious Diseases,185(9): 1277-. Bivalent II-TT and III-TT glycoconjugate vaccines (glycoconjugate vaccine) and methods of use including Ia-CRM197、Ib-CRM197And III-CRM197Trivalent vaccines for glycoconjugates have also been investigated (Baker JID 2003; clinical trials. gov NCT01193920, NCT01412801, and NCT 01446289). However, no GBS vaccine has been approved.
Furthermore, although the trivalent vaccine covered > 90% of invasive strains causing neonatal disease in south Africa (Madzivhandila, M., et al, PloS One,6(3): e17861(2011)), these same serotypes only represented 62% and 66% of invasive isolates in North America and Europe, respectively, according to recent monitoring of neonatal isolates of 901 global collections of samples collected from Tigecycline Evaluation and Surveillance Trial (T.E.S.T., http:// www.testsurveillance.com /) 2004 to 2013.
Analysis of the strains obtained from the t.e.s.t. samples showed that 95% of the collected strains were of one of the five major serotypes (Ia, Ib, II, III and V) recorded, while the other 3% were serotype IV. A series of publications have also identified the appearance of serotype IV in the past decade of America and Europe (Diedrick, M.J., et al, J.Clin. Microbiol.,48(9):3100-3104 (2010); Teatero (2014); Meehan, M.et al, European Journal of Clinical Microbiology & Infections, 33(7): 1155-. Studies investigating adult rectal/vaginal carriers (carriage), a risk factor for GBS transmission to infants, also found that 97% of isolates belong to one of these six serotypes, whereas serotype IV represents a frequency of-4%. This study was designed to monitor beta-hemolytic streptococci (GBS), Clostridium difficile (Clostridium difficile), and Staphylococcus aureus (Staphylococcus aureus) carriers of healthy adults in the united states (see Matson, m.a., et al, ICAAC, Abstract I-306(Washington, DC, sep.5-9,2014)).
Similarly, analysis of the T.E.S.T. sample showed that 98% of blood isolates from older US adults > 65 years of age belonged to the same six major serogroups. The most obvious difference between the elderly isolates and other populations is serogroup distribution. In terms of isolates from elderly patients, serotype V strains constitute the largest group (34%, 18% relative to neonatal or adult strains).
Other studies have found that there is regional variation in serotype prevalence. For example, the serotype VI and VIII isolates have been identified as the primary colonizers of healthy pregnant women in Japan (Colonizer) (Lachenauer, C.S., et al, JID 179(4):1030-1033 (1999)).
Thus, there is a need for polysaccharide-protein conjugate vaccines or monoclonal antibodies to confer passive immunity as a means to prevent or treat GBS disease (including those caused by the emerging serotype IV) in a large population worldwide.
Summary of The Invention
The present invention relates to novel immunogenic GBS polysaccharide-protein conjugates, methods of making the conjugates, and immunogenic compositions comprising the conjugates, and includes the inventions disclosed in U.S. provisional application No. 62/156,500, filed on 5/4/2015, U.S. provisional application No. 62/237,813, filed on 5/4/2015, and U.S. provisional application No. 62/237,820, filed on 6/10/2015, which are incorporated herein by reference in their entirety. The following items describe certain aspects and embodiments of the invention.
In one aspect, the invention relates to an immunogenic polysaccharide-protein conjugate comprising a capsular polysaccharide from Group B Streptococcus (GBS) and a carrier protein, wherein the capsular polysaccharide has a sialic acid level of greater than about 60%, greater than about 95%, or about 100%. In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (degree of sialylation (sialylation level) above about 60%). In another embodiment, the capsular polysaccharide is selected from the group consisting of: serotypes Ia, Ib, II, III, IV, V, VI, VII, VIII and IX.
In further aspects, the immunogenic polysaccharide-protein conjugate comprises a capsular polysaccharide having about 1.0mM sialic acid per mM polysaccharide, such as at least about 0.6, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, or 0.95mM sialic acid per mM polysaccharide.
In another aspect of the invention, the immunogenic conjugate comprises a capsular polysaccharide having a molecular weight of about 5kDa to about 1,000kDa, about 25kDa to about 750kDa, about 25kDa to about 400kDa, about 25kDa to about 200kDa, or about 100kDa to about 400 kDa.
In further embodiments, the immunogenic conjugate of the invention has a molecular weight of about 300kDa to about 20,000kDa, such as about 1,000kDa to about 15,000kDa, or about 1,000kDa to about 10,000 kDa.
In one embodiment, the immunogenic conjugate comprises a capsular polysaccharide that is about 0% to about 40% O-acetylated, such as less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% O-acetylated.
In one embodiment, the immunogenic conjugate comprises a capsular polysaccharide having at least about 0.1, 0.2, 0.3, 0.35, or about 0.4mM O-acetate per mM saccharide repeat unit. In another embodiment, the immunogenic conjugate comprises a capsular polysaccharide having less than about 0.01, 0.02, 0.03, 0.04, or 0.05mM O-acetate per mM saccharide repeat unit.
In one embodiment, the immunogenic conjugate comprises CRM197Or tetanus toxoid as the carrier protein. In a particular embodiment, the carrier protein is CRM197
Further aspects of the invention relate to methods of isolating capsular polysaccharides comprising reacting an organic reagent with a cell broth (cell broth) comprising a capsular polysaccharide producing bacterium. In one embodiment, the method further comprises a centrifugation step. In another embodiment, the method further comprises a filtration step. In particular embodiments, the capsular polysaccharide producing bacterium is selected from the group consisting of: streptococcus agalactiae (Streptococcus agalactiae), Streptococcus pneumoniae (Streptococcus pneumoniae), Staphylococcus aureus (Staphylococcus aureus), Neisseria meningitidis (Neisseria meningitidis), Escherichia coli (Escherichia coli), Salmonella typhi (Salmonella typhi), Haemophilus influenzae (Haemophilus influenzae), Klebsiella pneumoniae (Klebsiella pneumoniae), Enterococcus faecium (Enterococcus faecalis), and Enterococcus faecalis (Enterococcus faecalis). In one embodiment, the hydroxylamine is selected from the group consisting of the amines listed in table 2. In further embodiments, the hydroxylamine is selected from the group consisting of: dibenzylhydroxylamine (dibenzylhydroxylamine); diethylhydroxylamine (diethyl hydroxylamine); hydroxylamine (hydroxyimine); ethylenediamine (ethylenediamine); triethylenetetramine (triethylenetramine); 1,1,4,7,10,10 hexamethyltriethylenetetramine (1,1,4,7,10,10 hexamethylene triene tetramine); and 2,6,10, Trimethyl 2,6,10triazaundecane (2,6,10, trimethy 2,6,10 triazaundecane). In another embodiment, the concentration of hydroxylamine is from about 5mM to about 200 mM. In another embodiment, the pH of the reaction is from about 5.5 to about 9.5. In other embodiments, the reaction is carried out at a temperature of from about 20 ℃ to about 85 ℃. In another embodiment, the reaction time is from about 10 hours to about 90 hours.
In one aspect, the invention relates to an immunogenic composition comprising a polysaccharide-protein conjugate, wherein the conjugate comprises a capsular polysaccharide from Group B Streptococcus (GBS) serotype IV and at least one additional serotype selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In another embodiment, the conjugate comprises GBS serotype IV and at least two additional serotypes selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In another embodiment, the conjugate comprises GBS serotype IV and at least three additional serotypes selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In another embodiment, the conjugate comprises GBS serotype IV and at least four additional serotypes selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In a particular embodiment, the conjugate comprises capsular polysaccharides from serotypes Ia, Ib, II, III and IV. In another embodiment, the composition comprises GBS serotype V. In a particular embodiment, the conjugate comprises capsular polysaccharides from serotypes Ia, Ib, II, III, and V. In another embodiment, the immunogenic composition comprises six polysaccharide-protein conjugates, wherein the conjugates comprise capsular polysaccharides from group B streptococcus serotypes Ia, Ib, II, III, IV and V. One aspect of the invention relates to immunogenic compositions that do not have immune interference.
In one embodiment, the immunogenic composition further comprises a pharmaceutically acceptable excipient, buffer, stabilizer, adjuvant, cryoprotectant, salt, divalent cation, non-ionic detergent, free radical oxidation inhibitor, carrier, or mixture thereof. In other embodiments, a buffer is included. The buffer may be HEPES, PIPES, MES, Tris (trometamol), phosphate, acetate, borate, citrate, glycine, histidine, or succinate. In a preferred embodiment, the buffering agent is histidine.
In another embodiment, the immunogenic composition further comprises a surfactant. The surfactant may be polyoxyethylene sorbitan fatty acid ester, polysorbate-80, polysorbate-60, polysorbate-40, polysorbate-20, or polyoxyethylene alkyl ether (polyoxylethylene alkyl ether). In a preferred embodiment, the surfactant is polysorbate-80.
In another embodiment, the immunogenic composition further comprises an excipient. The excipient is selected from the group consisting of: starch, glucose, lactose, sucrose, trehalose (trehalose), raffinose (raffinose), stachyose (stachyose), melezitose (melezitose), dextran (dextran), mannitol, lactitol (lactitol), palatinit (palatinit), gelatin, malt, rice, flour, chalk (chalk), silica gel, sodium stearate, glycerol monostearate, talc, glycine, arginine, lysine, sodium chloride (NaCl), dried skim milk, glycerol, propylene glycol, water and ethanol. In a preferred embodiment, the excipient is sodium chloride.
In another embodiment, the immunogenic composition further comprises an adjuvant. In one such embodiment, the adjuvant is an aluminum-based adjuvant or QS-21. In a preferred embodiment, the adjuvant is selected from the group consisting of: aluminum phosphate, aluminum hydroxyphosphate, and aluminum hydroxide. In a more preferred embodiment, the adjuvant is aluminum phosphate.
In one aspect of the invention, the immunogenic composition comprises a buffer, a surfactant, an excipient, and optionally an adjuvant, wherein the composition is buffered to a pH of about 6.0 to about 7.0. In another aspect, an immunogenic composition comprises histidine, polysorbate-80, sodium chloride, and optionally aluminum phosphate, wherein the composition is buffered to a pH of about 6.0 to about 7.0. In a preferred embodiment, the immunogenic composition comprises histidine in a range of about 10mM to about 25mM, polysorbate-80 in a range of about 0.01% to about 0.03% (v/w), sodium chloride (NaCl) in a range of about 10mM to about 250mM, and optionally aluminum as aluminum phosphate in a range of about 0.25mg/ml to about 0.75 mg/ml. In another aspect of the invention, the immunogenic composition comprises a dose of about 5mcg/ml to about 50 mcg/ml.
In another aspect of the invention, the immunogenic composition is lyophilized, optionally in the presence of at least one excipient. In one embodiment, the at least one excipient is selected from the group consisting of: starch, glucose, lactose, sucrose, trehalose, raffinose, stachyose, melezitose, dextran, mannitol, lactitol, arabitol, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, glycine, arginine, lysine, sodium chloride (NaCl), dried skim milk, glycerol, propylene glycol, water and ethanol. In a preferred embodiment, the at least one excipient is selected from the group consisting of: sucrose, mannitol, and glycine. In a particular embodiment, the at least one excipient is sucrose. In one aspect, the lyophilized composition comprises from about 1% (w/v) to about 10% (w/v) of at least one excipient, preferably more than about 5.5% (w/v). In another embodiment, the freeze-dried composition comprises additional excipients. In one such embodiment, the additional excipient is mannitol or glycine. In a preferred embodiment, the freeze-dried composition comprises from about 1% (w/v) to about 10% (w/v) of additional excipients. In another embodiment, the freeze-dried composition is reconstituted with water, water for injection (WFI), adjuvant suspension, or saline. In particular embodiments, the diluent is a suspension of any adjuvant described herein, such as an aluminum-based adjuvant suspension, preferably an aluminum phosphate suspension.
Another aspect of the invention relates to a method of eliciting an immune response to GBS comprising administering to a subject an effective amount of an immunogenic composition as described herein. In one embodiment, the present invention relates to a method of preventing or alleviating a disease or condition associated with GBS in a subject comprising administering to the subject an effective amount of an immunogenic composition as described herein. In particular embodiments, the subject is a female intended to be pregnant or a pregnant female. In one such embodiment, a pregnant female is in its second half of pregnancy (second half of pregnancy), such as at least 20 weeks of gestation or at least 27 weeks of gestation. In a preferred embodiment, the pregnant female is between 27 and 36 weeks gestation. In another embodiment, the subject is an older adult, such as an adult of 50 years or older, an adult of 65 years or older, and an adult of 85 years or older. In another embodiment, the subject is immunocompromised. In one aspect, the subject may have a medical condition selected from the group consisting of: obesity, diabetes, HIV infection, cancer, cardiovascular disease, or liver disease. In a preferred embodiment, the group B Streptococcus is Streptococcus agalactiae (Streptococcus agalactiae).
A further aspect of the invention relates to antibodies that bind to the capsular polysaccharide in the immunogenic composition of the invention. In some embodiments, the antibody is produced when the immunogenic composition is administered to a subject. In another aspect, to compositions comprising the antibodies of the invention.
Another aspect of the invention relates to a method of conferring passive immunity (passive immunity) to a subject, comprising the steps of: (a) generating an antibody preparation using the immunogenic composition described herein; and (b) administering the antibody formulation to the subject to confer passive immunity.
One aspect of the present invention relates to a method of making an immunogenic polysaccharide-protein conjugate of the invention comprising the steps of: (a) reacting a GBS capsular polysaccharide with an oxidizing agent to produce an activated polysaccharide; and (b) reacting the activated polysaccharide with a carrier protein to produce a polysaccharide-protein conjugate, wherein step (b) is carried out in a polar aprotic solvent. The solvent may be dimethyl sulfoxide (DMSO), sulfolane, Dimethylformamide (DMF), or Hexamethylphosphoramide (HMPA). In a preferred embodiment, the solvent is dimethyl sulfoxide (DMSO).
In one embodiment, the polysaccharide is reacted with 0.01 to 10.0 molar equivalents of an oxidizing agent. In a particular embodiment, the oxidizing agent is periodate. In one such embodiment, the periodate salt is sodium periodate.
In another embodiment, the oxidation reaction is from 1 hour to 50 hours. In another embodiment, the temperature of the oxidation reaction is maintained at about 2 ℃ to about 25 ℃. In another embodiment, the oxidation reaction is carried out in a buffer selected from the group consisting of: sodium phosphate, potassium phosphate, 2- (N-morpholino) ethanesulfonic acid (MES) and Bis-Tris. In one such embodiment, the buffer is at a concentration of about 1mM to about 500 mM. In a particular embodiment, the oxidation reaction is carried out at a pH of from about 4.0 to about 8.0.
In another aspect of the invention, the oxidizing agent is 2,2,6,6-tetramethyl-1-piperidinyloxy (2,2,6,6-tetramethyl-1-piperidinyloxy) (TEMPO). In one such embodiment, N-chlorosuccinimide (NCS) is a co-oxidant.
In one embodiment, step (a) of the present invention of making the immunogenic polysaccharide-protein conjugate further comprises quenching the oxidation reaction by adding a quencher.
In another embodiment, the concentration of polysaccharide is from about 0.1mg/mL to about 10.0 mg/mL.
In a further embodiment, the activated polysaccharide has a Degree of Oxidation (DO) of 5 to 25.
In another aspect of the invention, the method further comprises the step of freeze drying the activated polysaccharide. In one embodiment, the activated polysaccharide is freeze-dried in the presence of a sugar selected from the group consisting of: sucrose, trehalose, raffinose, stachyose, melezitose, dextran, mannitol, lactitol, and xylitol.
In another aspect of the present invention, step (b) of the present invention of making an immunogenic polysaccharide-protein conjugate comprises: (1) mixing the activated polysaccharide with a carrier protein, and (2) reacting the mixed activated polysaccharide and carrier protein with a reducing agent to form a GBS capsular polysaccharide-carrier protein conjugate. In one embodiment, the concentration of activated polysaccharide in step (2) is from about 0.1mg/mL to about 10.0 mg/mL. In a further embodiment, the initial ratio (weight/weight) of activated polysaccharide to carrier protein is5:1 to 0.1: 1. In another embodiment, the reducing agent is selected from the group consisting of: sodium cyanoborohydride (NaBH)3CN), sodium triacetoxyborohydride, sodium borohydride and zinc borohydride in the presence of Bronsted acid or Lewis acid, amine boranes such as pyridine borane, 2-picoline borane, 2, 6-diborane-methanol, dimethylamine-borane, t-BuMeiPrN-BH3benzylamine-BH3Or 5-ethyl-2-picoline borane (PEMB). In a preferred embodiment, the reducing agent is (NaBH)3CN). In another embodiment, the amount of reducing agent is from about 0.1 to about 10.0 molar equivalents. In another embodiment, the duration of the reduction reaction of step (2) is from 1 hour to 60 hours. In another embodiment, the temperature of the reduction reaction is maintained between 10 ℃ and 40 ℃.
In a further aspect of the invention, the method of making an immunogenic polysaccharide-protein conjugate further comprises the step of capping unreacted aldehydes by adding borohydride (step (c)). In one embodiment, the amount of borohydride is from about 0.1 to about 10.0 molar equivalents. In another embodiment, the borohydride is selected from the group consisting of: sodium borohydride (NaBH)4) Sodium cyanoborohydride, lithium borohydride, potassium borohydride, tetrabutylammonium borohydride, calcium borohydride and magnesium borohydride. In a preferred embodiment, the borohydride is sodium borohydride (NaBH)4). In another embodiment, the duration of the capping step is from 0.1 hour to 10 hours. In another embodiment, the temperature of the capping step is maintained at about 15 ℃ to about 45 ℃.
In another aspect of the invention, the method further comprises the step of purifying the polysaccharide-protein conjugate. In one embodiment, the polysaccharide-protein conjugate comprises less than about 40% free polysaccharide compared to the total amount of polysaccharide. In another embodiment, the ratio of polysaccharide to carrier protein in the conjugate (weight/weight) is from about 0.5 to about 3.0. In another embodiment, the degree of conjugation of the conjugate is from 2 to 15.
In another aspect the invention relates to the manufacture of the immunogenic polysaccharide-protein of the inventionA method of mass conjugation comprising the steps of: (a) reacting the isolated GBS capsular polysaccharide with an oxidizing agent; (b) quenching the oxidation reaction of step (a) by adding a quenching agent to produce an activated GBS capsular polysaccharide; (c) mixing the activated GBS capsular polysaccharide with a carrier protein, (d) reacting the mixed activated GBS capsular polysaccharide and carrier protein with a reducing agent to form a GBS capsular polysaccharide-carrier protein conjugate, and (e) by adding sodium borohydride (NaBH)4) Capping the unreacted aldehyde, wherein steps (c), (d) and (e) are performed in DMSO.
Brief Description of Drawings
FIG. 1 comparison from GBS Ia-CRM197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197Serum from mice immunized with a monovalent vaccine and opsonin activity of isolated igg (isolated igg).
FIG. 2GBS Ia-CRM197Stability after accelerated storage (4 weeks) at 50 ℃ (as shown by% change in molecular weight, using SEC MALLS).
FIG. 3GBS Ib-CRM197Stability after accelerated storage (4 weeks) at 50 ℃ (SEC MALLS was used as shown by% change in molecular weight).
FIG. 4GBS II-CRM197Stability after accelerated storage (4 weeks) at 50 ℃ (SEC MALLS was used as shown by% change in molecular weight).
FIG. 5GBS III-CRM197Stability after accelerated storage (4 weeks) at 50 ℃ (SEC MALLS was used as shown by% change in molecular weight).
FIG. 6GBS IV-CRM197Stability after accelerated storage (4 weeks) at 50 ℃ (SEC MALLS was used as shown by% change in molecular weight).
FIG. 7GBS V-CRM197Stability after accelerated storage (4 weeks) at 50 ℃ (SEC MALLS was used as shown by% change in molecular weight).
FIG. 8GBS Ia-CRM197、GBS Ib-CRM197、GBS III-CRM197And GBS IV-CRM197Stability after storage at 37 ℃ (as shown by free sialic acid).
FIG. 9 hexavalent GBS vaccine (GBS Ia-CRM) using succinate as buffer197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) Stability of pH of (1).
FIG. 10 hexavalent GBS vaccine using histidine as a buffer (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) Stability of pH of (1).
FIG. 11 hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) The effect of histidine buffer concentration in (c) on binding of GBS conjugate to aluminum (dose of 10 mcg/ml).
FIG. 12 hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) The effect of histidine buffer concentration in (c) on binding of GBS conjugate to aluminum (dose 40 mcg/ml).
FIG. 13 hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) Effect of polysorbate-80 concentration on total antigenicity loss at stirring pressure%.
FIG. 14 hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) The effect of aluminum concentration in GBS conjugates on aluminum binding.
FIG. 1510 mcg/ml dose of freeze-dried hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) 5.5% (w/v) sucrose in (C) antigenicity recovery for each serotypeThe effects of the complex.
FIG. 1610 mcg/ml dose of freeze-dried hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) 7.0% (w/v) sucrose in (1) on the recovery of antigenicity of each serotype.
FIG. 1710 mcg/ml dose of freeze-dried hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) 8.5% (w/v) sucrose in (1) on the recovery of antigenicity of each serotype.
FIG. 1850 mcg/ml dose of freeze-dried hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) 5.5% (w/v) sucrose in (1) on the recovery of antigenicity of each serotype.
FIG. 1950 mcg/ml dose of freeze-dried hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) 7.0% (w/v) sucrose in (1) on the recovery of antigenicity of each serotype.
FIG. 2050 mcg/ml dose of freeze-dried hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) 8.5% (w/v) sucrose in (1) on the recovery of antigenicity of each serotype.
FIG. 2140 mcg/ml dose of freeze-dried hexavalent GBS vaccine (GBS Ia-CRM) 197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) 7.0% (w/v) sucrose in (1) on the recovery of antigenicity of each serotype.
FIG. 2240 mcg/ml dose of freeze-dried hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) 2.0% (w/v) sucrose and 4.0% (w/v) mannitol in (C) on the recovery of antigenicity of each serotype.
FIG. 2340 mcg/ml dose of freeze-dried hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) 3.0% (w/v) sucrose and 3.0% (w/v) mannitol in (C) on the recovery of antigenicity of each serotype.
FIG. 2440 mcg/ml dose of freeze-dried hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) 2.0% (w/v) sucrose and 4.0% (w/v) glycine in (C) on the recovery of antigenicity of each serotype.
FIG. 2540 mcg/ml dose of freeze-dried hexavalent GBS vaccine (GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) 3.0% (w/v) sucrose and 3.0% (w/v) glycine in (C) on the recovery of antigenicity of each serotype.
Detailed Description
It is to be understood that this invention is not limited to the particular methodology and experimental conditions described, as such methodologies and conditions may vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.
Although any methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, the preferred methods and materials are now described. All publications mentioned herein are incorporated herein by reference in their entirety.
Terms used herein have meanings that are understood and known by those skilled in the art, however, for convenience and completeness, specific terms and their meanings are set forth below and throughout the specification.
As used in this specification and the appended claims, the singular forms "a", "an", and "the" include plural referents unless the context clearly dictates otherwise. Thus, for example, reference to "the method" includes one or more methods and/or steps of the type described herein, and/or those that would be apparent to one skilled in the art upon reading this disclosure, and so forth.
The term "about" or "approximately" means within a statistically significant range of numerical values. Such a range may be within an order of magnitude of a given value or range, typically within 20%, more typically within 10%, and even more typically within 5%. The allowable variations encompassed by the terms "about" or "approximately" depend on the particular system under study and can be readily understood by one skilled in the art. Whenever a range is recited in this application, each integer within the range is also contemplated as an embodiment of the invention.
It is to be noted that, in the present disclosure, terms such as "comprising", "including", "containing" and the like may have meanings given to them in U.S. patent law; for example, it may mean "including" and the like. Such terms are intended to encompass a particular ingredient or group of ingredients, but not to exclude any other ingredient. Terms such as "consisting essentially of" have the meaning attributed to them in U.S. patent law, e.g., it allows the inclusion of additional ingredients or steps that do not detract from the novelty or essential features of the invention, i.e., it excludes additional unrecited ingredients or steps that detract from the novelty or essential features of the invention, and it excludes ingredients or steps of the prior art (such as documents cited or incorporated by reference herein), especially the objects of this document are to define embodiments that are patentable (e.g., are novel, nonobvious, inventive compared to the prior art (e.g., compared to documents cited or incorporated by reference herein). Also, the term "consisting of has the meaning attributed to them in U.S. patent law; that is, these terms are closed. Thus, these terms are meant to include a particular ingredient or group of ingredients and to exclude all other ingredients.
The term "antigen" generally refers to a biomolecule, typically a protein, peptide, polysaccharide, lipid or conjugate, containing at least one epitope to which a cognate antibody can selectively bind; or in some cases, "antigen" refers to an immunogenic substance that stimulates production of antibodies or a T cell response, or both, in an animal, including compositions that are injected or absorbed into an animal. The immune response may be raised to the entire molecule, or to one or more different parts of the molecule (e.g., an epitope or hapten). The term may be used to refer to a homogeneous or heterogeneous population of individual molecules, or antigenic molecules. Antigens may be recognized by antibodies, T cell receptors, or other components of specific humoral and/or cellular immunity. The term "antigen" includes all relevant antigenic epitopes. Epitopes of a given antigen can be identified using any number of Epitope Mapping techniques well known in the art (see, e.g., Epitope Mapping Protocols in Methods in Molecular Biology, vol.66(Glenn E. Morris, Ed.,1996) Humana Press, Totowa, NJ). For example, linear epitopes can be determined by, for example: simultaneously synthesizing a plurality of peptides (such peptides corresponding to portions of the protein molecule) on a solid support, and reacting such peptides with an antibody while such peptides are still attached to the support. Such techniques are known in the art and are described, for example, in U.S. Pat. nos. 4,708,871; geysen, H.M., et al, Proc.Natl.Acad.Sci.USA,81: 3998-; geysen, H.M., et al, mol. Immunol.,23(7):709-715(1986), the entire contents of which are incorporated herein by reference. Similarly, conformational epitopes can be identified by determining the spatial configuration of amino acids, such as by, for example, x-ray crystallography and 2-dimensional nuclear magnetic resonance (see, e.g., Epitope Mapping Protocols, supra). Furthermore, for the purposes of the present invention, "antigen" may also be used to refer to a protein that includes modifications, such as deletions, insertions, and substitutions (typically conservative in nature, but which may be non-conservative) of the native sequence, so long as the protein retains the ability to elicit an immune response. These modifications may be deliberate, as by site-directed mutagenesis, or by specific synthetic procedures, or by genetic engineering methods, or may be accidental, such as by mutation of the host producing the antigen. Furthermore, the antigen may be derived, obtained, or isolated from a microorganism (e.g., a bacterium), or may be the entire organism. Similarly, oligonucleotides or polynucleotides that express antigens (such as in nucleic acid immunization applications) are also included in this definition. Also included are synthetic antigens such as polyepitopes, flanking epitopes and other recombinantly or synthetically derived antigens (Bergmann, C., et al, Eur. J. Immunol.,23(11): 2777-.
The terms "vaccine" or "vaccine composition" (which are used interchangeably) refer to a pharmaceutical composition that includes at least one immunogenic composition that can elicit an immune response in an animal.
Capsular polysaccharide
The term "saccharide" as used herein refers to a monosaccharide moiety or monosaccharide unit, and a combination of two or more monosaccharide moieties or monosaccharide units covalently linked to form a disaccharide, an oligosaccharide, and a polysaccharide. The term "sugar" is used interchangeably with the term "carbohydrate". The polysaccharide may be linear or branched.
As used herein, "monosaccharide" refers to a monosaccharide residue in an oligosaccharide. The term "disaccharide" as used herein refers to a polysaccharide consisting of two monosaccharide units or moieties linked together by glycosidic bonds.
In one embodiment, the polysaccharide is an Oligosaccharide (OS). As used herein, "oligosaccharide" refers to a compound containing two or more monosaccharide units or moieties. In the context of oligosaccharides, an individual monomeric unit or moiety is a monosaccharide that is (or can be) bound to another monosaccharide unit or moiety through a hydroxyl group. Oligosaccharides can be prepared from protected mono-residue sugars by chemical synthesis or by chemical degradation of biologically produced polysaccharides. Alternatively, the oligosaccharides may be produced by an in vitro enzymatic process.
In a preferred embodiment, the polysaccharide is a Polysaccharide (PS), which refers to a linear or branched polymer having at least 5 monosaccharide units or moieties. For clarity, a larger number of repeating units (where n is greater than about 5, such as greater than about 10) will be referred to herein as polysaccharides.
In one embodiment, the polysaccharide is a cell surface polysaccharide. Cell surface polysaccharides refer to polysaccharides which are at least partially located on the outermost bacterial cell membrane or bacterial cell surface (including peptidoglycan layer, cell wall and capsule). Typically, cell surface polysaccharides are associated with the induction of immune responses in vivo. The cell surface polysaccharide may be a "cell wall polysaccharide" or a "capsular polysaccharide". Cell wall polysaccharides typically form a discontinuous layer on the surface of bacteria.
In one embodiment, the polysaccharide is a capsular polysaccharide. Capsular polysaccharide refers to a carbohydrate polymer comprising one or more monosaccharide repeating units linked by glycosidic linkages. Capsular polysaccharides typically form a capsular film-like layer around the bacterial cells. "capsular polysaccharide" refers to the polysaccharide capsule outside the cell wall of most streptococcus isolates. For example, all GBS capsular polysaccharides have a branched chain type repeat structure with a terminal α 2-3 linked sialic acid required for bacterial virulence. Pod-associated sialic acid (quantified by HPLC analysis) was detected in > 94% of invasive neonatal isolates from t.e.s.t. cultured in vitro.
The present inventors have found that the sialic acid level of the GBS capsular polysaccharide is an important feature in generating an immune response. The prior art provides only conflicting information on sialic acid levels of serotype V, and it was found that desialylated serotype V is preferred (international patent application publication No. WO 2012/035519) and that serotype V with a sialic acid content > 50% can be used (international patent application publication No. WO 2014/053612). However, none of these references describe the importance of sialic acid levels on the immunogenicity of at least a large proportion of GBS polysaccharides. The present inventors have surprisingly found that GBS capsular polysaccharides require about 60% or more sialic acid prior to conjugation to provide an immune response comparable to those polysaccharides with native sialic acid levels (i.e., 100% or more than about 95%). Even a sialic acid level of 58% (within the previously disclosed range for serotype V) negatively affected immunogenicity.
Thus, in one embodiment of the invention, the capsular polysaccharide comprises its natural sialic acid level (such as about 100% or more than about 95%). In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (sialylation degree above about 60%), such as up to about 35% (sialylation degree above about 65%), up to about 30% (sialylation degree above about 70%), up to about 25% (sialylation degree above about 75%), up to about 20% (sialylation degree above about 80%), up to about 15% (sialylation degree above about 85%), up to about 10% (sialylation degree above about 90%), and up to about 5% (sialylation degree above about 95%).
It should be noted that a sialic acid level of 100% corresponds to about 1.0mM sialic acid per mM polysaccharide. Thus, the capsular polysaccharide may have about 1.0mM sialic acid per mM of polysaccharide, such as at least about 0.95mM sialic acid per mM of polysaccharide. In another embodiment, the capsular polysaccharide may have at least about 0.6mM sialic acid per mM of polysaccharide, such as at least about 0.65mM sialic acid per mM of polysaccharide, at least about 0.7mM sialic acid per mM of polysaccharide, at least about 0.75mM sialic acid per mM of polysaccharide, at least about 0.8mM sialic acid per mM of polysaccharide, at least about 0.85mM sialic acid per mM of polysaccharide, at least about 0.9mM sialic acid per mM of polysaccharide, or at least about 0.95mM sialic acid per mM of polysaccharide.
The terminal sialic acid residues of certain Capsular Polysaccharide (CP) serotypes are partially O-acetylated (OAc) (Lewis, A.L., et al, Proceedings of the National Academy of Sciences USA,101(30):11123-8 (2004)). Serotypes Ib, III, IV, V, VI and IX were partially O-acetylated (up to-40%), whereas serotypes Ia, II and VII were only minimally or non-O-acetylated (less than about 5%) (Lewis 2004). In one embodiment of the invention, the capsular polysaccharide comprises its natural O-acetylation level (about 0% to about 40%). In another embodiment, the capsular polysaccharide may be de-O-acetylated (less than about 5%). The degree of O-acetylation of a polysaccharide or oligosaccharide can be determined by any method known in the art, for example by proton NMR (Lemercinier, X., et al, Carbohydrate Research,296:83-96 (1996); Jones, C., et al, Journal of Pharmaceutical and biological Analysis,30:1233-1247 (2002); International patent application publication Nos. WO 2005/033148 and WO 00/56357). Another commonly used method is described by Hestrin, S., J.biol.chem.,180:249-261 (1949).
It should also be noted that 100% O-acetate corresponds to about 1.0mM O-acetate per mM sugar repeat unit. Thus, the partially O-acetylated polysaccharide comprises at least about 0.1, 0.2, 0.3, 0.35, or about 0.4mM O-acetate per mM saccharide repeat unit. The de-O-acetylated polysaccharide comprises less than about 0.01, 0.02, 0.03, 0.04, or 0.05mM O-acetate per mM saccharide repeat unit.
Streptococcal microorganisms that can cause invasive disease are also generally capable of producing CP that coats the bacteria and enhances their resistance to elimination by the host's innate immune system. CP serves to shield bacterial cells in a protective capsule that renders the bacteria resistant to phagocytosis and intracellular killing. Bacteria lacking a capsule are more susceptible to phagocytosis. Capsular polysaccharides are often important virulence factors (viral factors) of many bacterial pathogens, including Haemophilus influenzae (Haemophilus influenzae), Streptococcus pneumoniae (Streptococcus pneumoniae), Neisseria meningitidis (Neisseria meningitidis) and Staphylococcus aureus (Staphylococcus aureus).
Capsular polysaccharides may be used to serotype a particular species. Typing is usually accomplished by reaction with specific antisera or monoclonal antibodies raised against specific structures or unique epitope characteristics of the capsular polysaccharide. There are ten GBS serotypes: ia. Ib and II to IX (Ferrieri, P., et al, emery. Infect. Dis. [ Internet ],19(4) (2013), available under http:// wwwnc. cdc. gov/eid/article/19/4/12-1572_ article).
In one embodiment of the invention, the polysaccharide is isolated from Streptococcus agalactiae (Streptococcus agalactiae). Polysaccharides can be isolated from any of the coated strains of Streptococcus agalactiae (S.agalactiae) (encapsed strain), such as 090, A909(ATCC accession number BAA-1138), 515(ATCC accession number BAA-1177), B523, CJB524, MB 4052(ATCC accession number 31574), H36B (ATCC accession number 12401), S40, S42, MB 4053(ATCC accession number 31575), M709, 133, 7357, PFEGBST0267, MB 4055(ATCC accession number 31576), 18RS21(ATCC accession number BAA-1175), S16, S20, V8(ATCC accession number 12912973), 539 21, DK23, UAB, 5401, PFEGBST0708, MB 4082(ATCC accession number 31577), M132, NI, M781(ATCC accession number BAA-22), PF1241242 (ATCC accession number 1243), ATCC accession number 4903, ATCC accession number 3195, ATCC accession number 3172, ATCC accession number 3173, ATCC accession number 7342, ATCC accession number 3172, ATCC accession number 3173, ATCC accession number 7346, ATCC accession number 3139, ATCC accession number 3172, ATCC accession number 3173, ATCC accession number 7346, ATCC accession number 31577, ATCC accession number 437817, ATCC accession number 7813, ATCC accession number 3172, ATCC accession number 7819, ATCC accession number 7815, ATCC accession number 7819, ATCC accession number 7815, ATCC accession number 7819, ATCC accession number 7815, ATCC accession number 7819, ATCC accession number 7815, ATCC accession number 7819, ATCC accession number 7815, ATCC accession number 7819, ATCC accession number 7815, ATCC accession number 7819, ATCC accession, CJB111(ATCC accession number BAA-23), CJB112, 2603V/R (ATCC accession number BAA-611), NCTC 10/81, CJ11, PFEGBST0837, 118754, 114852, 114862, 114866, 118775, B4589, B4645, SS1214, CZ-PW-119, 7271, CZ-PW-045, JM9130013, 9130672, IT-NI-016, IT-PW-62 and IT-PW-64.
The polysaccharides described herein can be isolated by methods known in the art, including, for example, the methods described herein. As used herein, "isolated" refers to obtained from and isolated from a particular source. The term "isolated" further refers to not being in its respective naturally occurring form, state, and/or environment. For example, "isolated from Streptococcus" refers to material obtained from and isolated from Streptococcus cells. Isolated polysaccharides are not naturally occurring. The term "isolated" refers to the removal of the substance from its original environment (e.g., from the natural environment if it is naturally occurring, or from its host organism if it is a recombinant entity, or from one environment to a different environment). For example, an "isolated" capsular polysaccharide, protein, or peptide is substantially free of cellular material or other contaminating proteins (from the cell or tissue source from which the protein is derived), or when it is chemically synthesized, it is substantially free of chemical precursors or other chemicals, or is otherwise present in the mixture as part of a chemical reaction. In the present invention, the protein or polysaccharide may be isolated from the bacterial cells or from cell debris so that it is provided in a form that can be used in the manufacture of an immunogenic composition. The term "isolated" or "isolating" may include purification, including methods known in the art for purifying isolated polysaccharides and/or methods described herein. "substantially free of cellular material" includes preparations of the polypeptide/protein in which the polypeptide/protein is separated from cellular components of the cell from which it is isolated or recombinantly produced. Thus, a capsular polysaccharide, protein or peptide that is substantially free of cellular material includes preparations of capsular polysaccharides, proteins or peptides having less than about 30%, 20%, 10%, 5%, 2.5%, or 1% (by dry weight) contaminating protein or polysaccharide or other cellular material. When the polypeptide/protein is recombinantly produced, it is also preferably substantially free of culture medium, i.e., the protein preparation comprises less than about 20%, 10%, or 5% by volume of culture medium. When the polypeptide/protein or polysaccharide is made by chemical synthesis, it is preferably substantially free of chemical precursors or other chemicals, i.e., it is separate from the chemical precursors or other chemicals involved in synthesizing the protein or polysaccharide. Thus, such preparations of polypeptides/proteins or polysaccharides have less than about 30%, 20%, 10%, 5% (by dry weight) of chemical precursors or compounds of non-target polypeptide/protein or polysaccharide fragments.
In one embodiment of the invention, the polysaccharide is isolated from bacteria. In another embodiment of the invention, the polysaccharide is produced recombinantly. In another embodiment, the polysaccharide is synthesized according to conventional methods or chemically. In another embodiment of the invention, the polysaccharide is produced by cloning and expressing the biosynthetic pathway that produces the polysaccharide followed by expression in an alternative host. In one embodiment, the polysaccharide is immunogenic. For example, the inventors of the present application have found that each of the polysaccharides described herein is capable of eliciting or eliciting an immune response. The term "immunogenic" refers to the ability to initiate, trigger, elicit, enhance, ameliorate, and/or amplify a humoral and/or cell-mediated immune response in a mammal. In one embodiment, the mammal is a human, primate, rabbit, pig, mouse, and the like.
The molecular weight of the capsular polysaccharide is one consideration for immunogenic compositions. High molecular weight capsular polysaccharides are capable of eliciting certain antibody immune responses due to the higher valency of the epitopes present on the surface of the antigen. Isolation and purification of high molecular weight capsular polysaccharides are contemplated for use in the conjugates, compositions and methods of the invention.
However, in one embodiment, the size of the polysaccharide may be altered to a Molecular Weight (MW) range that is less than the molecular weight of the native capsular polysaccharide (prior to conjugation with the carrier protein). The purified capsular polysaccharide is reduced in size to produce a conjugate having advantageous filterability characteristics and/or yields.
In one such embodiment, the purified capsular polysaccharide is reduced in size by high pressure homogenization. High pressure homogenization achieves high shear rates by pumping the process stream through a flow path of sufficiently small size. Shear rate is increased by using a larger applied homogenization pressure, while exposure time may be increased by recycling the feed stream through the homogenizer.
In one embodiment, the polysaccharide described herein is capable of inducing opsonin activity. In another embodiment, the polysaccharides described herein are capable of inducing opsonin activity and phagocytic activity (e.g., opsonophagocytic activity).
Opsonin activity or opsonization refers to the process of binding an opsonin (e.g., an antibody or complement factor) to an antigen (e.g., an isolated polysaccharide described herein), which facilitates the attachment of the antigen to phagocytic or phagocytic cells (e.g., macrophages, dendritic cells, and polymorphonuclear leukocytes (PMNL)). Certain bacteria (such as, for example, coated bacteria that are not normally phagocytized due to the presence of a capsule) may become more readily recognized by phagocytic cells when coated with opsonin antibodies. In one embodiment, the polysaccharide elicits an immune response, such as, for example, an antibody, which is opsonin. In one embodiment, the opsonin activity is against a gram-positive coccus, preferably against a streptococcal species, more preferably against at least one strain of streptococcus agalactiae.
In another embodiment, the polysaccharide described herein is capable of eliciting a bactericidal immune response. In one embodiment, the bactericidal activity is against gram-positive cocci, preferably against a streptococcal species, more preferably against at least one strain of streptococcus agalactiae.
Methods for measuring opsonization, phagocytosis, and/or bactericidal activity are known in the art, such as, for example, by measuring a reduction in bacterial load in vivo (e.g., by measuring bacteremia levels in a mammal challenged with a streptococcal species) and/or by measuring bacterial cell killing in vitro (e.g., an in vitro opsonophagocytosis assay). In one embodiment, the polysaccharide is capable of inducing opsonin, phagocytosis, and/or bactericidal activity compared to an appropriate control group (such as, for example, compared to antisera raised against a gram-positive coccus killed by heat).
Serotype Ia
One embodiment comprises a serotype la GBS capsular polysaccharide. The structure of serotype Ia can be described as follows:
Figure BDA0003350538080000231
the serotype Ia capsular polysaccharide, prior to conjugation, has a molecular weight of about 5kDa to about 1,000kDa, such as about 25kDa to about 750kDa, about 25kDa to about 500kDa, about 25kDa to about 450kDa, about 25kDa to about 400kDa, about 25kDa to about 350kDa, about 25kDa to about 300kDa, about 25kDa to about 250kDa, about 25kDa to about 200kDa, about 50kDa to about 750kDa, about 50kDa to about 500kDa, about 50kDa to about 450kDa, about 50kDa to about 400kDa, about 50kDa to about 350kDa, about 50kDa to about 300kDa, about 50kDa to about 250kDa, about 50kDa to about 200kDa, about 75kDa to about 750kDa, about 75kDa to about 500kDa, about 75kDa to about 450kDa, about 75 to about 400kDa, about 75kDa to about 350kDa, about 75kDa to about 300kDa, about 75kDa to about 250kDa, about 75kDa, about 100 to about 200kDa, about 100 to about 100kDa, about 100kDa to about 300kDa, about 75kDa, About 100kDa to about 500kDa, about 100kDa to about 450kDa, about 100kDa to about 400kDa, about 100kDa to about 350kDa, about 100kDa to about 300kDa, about 200kDa to about 750kDa, about 200kDa to about 700kDa, about 200kDa to about 650kDa, about 200kDa to about 600kDa, about 200kDa to about 550kDa, about 200kDa to about 500kDa, about 200kDa to about 450kDa, about 200kDa to about 400kDa, about 250kDa to about 750kDa, about 250kDa to about 700kDa, about 250kDa to about 650kDa, about 250kDa to about 600kDa, about 250kDa to about 550kDa, about 250kDa to about 500kDa, about 250kDa to about 450kDa, about 250kDa to about 400kDa, about 300kDa to about 750, about 300kDa to about 700kDa, about 300kDa to about 650kDa, about 300kDa to about 600kDa, about 300kDa to about 550kDa, or about 300 to about 500 kDa. In a preferred embodiment, the capsular polysaccharide, prior to conjugation, has a molecular weight of from about 25kDa to about 200 kDa. In another preferred embodiment, the capsular polysaccharide, prior to conjugation, has a molecular weight of from about 100kDa to about 400 kDa. Any integer within any of the above ranges is contemplated as an embodiment of the present disclosure.
In a particular embodiment, the native GBS capsular polysaccharide serotype Ia is reduced in size using a high pressure homogenization process, but retains the structural features of the polysaccharide, such as sialic acid.
In one embodiment of the invention, the serotype Ia capsular polysaccharide comprises its natural sialic acid level, such as about 100% or above about 95%. In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (degree of sialylation greater than about 60%), such as up to about 35% (degree of sialylation greater than about 65%), up to about 30% (degree of sialylation greater than about 70%), up to about 25% (degree of sialylation greater than about 75%), up to about 20% (degree of sialylation greater than about 80%), up to about 15% (degree of sialylation greater than about 85%), up to about 10% (degree of sialylation greater than about 90%), or up to about 5% (degree of sialylation greater than about 95%) prior to conjugation.
In another embodiment, the serotype Ia capsular polysaccharide has about 1.0mM sialic acid per mM polysaccharide, such as at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation. In another embodiment, the capsular polysaccharide may have at least about 0.6mM sialic acid per mM polysaccharide, such as at least about 0.65mM sialic acid per mM polysaccharide, at least about 0.7mM sialic acid per mM polysaccharide, at least about 0.75mM sialic acid per mM polysaccharide, at least about 0.8mM sialic acid per mM polysaccharide, at least about 0.85mM sialic acid per mM polysaccharide, at least about 0.9mM sialic acid per mM polysaccharide, or at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation.
The serotype Ia capsular polysaccharide is less than about 5% O-acetylated. Some exemplary strains of serotype Ia capsular polysaccharide of the invention include 090, A909(ATCC accession number BAA-1138), 515(ATCC accession number BAA-1177), B523, CJB524 and MB 4052(ATCC accession number 31574).
Serotype Ib
One embodiment comprises a serotype Ib GBS capsular polysaccharide. The structure of serotype Ib can be described as follows:
Figure BDA0003350538080000261
the serotype Ib capsular polysaccharide, prior to conjugation, has a molecular weight of about 5kDa to about 1,000kDa, such as about 25kDa to about 750kDa, about 25kDa to about 500kDa, about 25kDa to about 450kDa, about 25kDa to about 400kDa, about 25kDa to about 350kDa, about 25kDa to about 300kDa, about 25kDa to about 250kDa, about 25kDa to about 200kDa, about 50kDa to about 750kDa, about 50kDa to about 500kDa, about 50kDa to about 450kDa, about 50kDa to about 400kDa, about 50kDa to about 350kDa, about 50kDa to about 300kDa, about 50kDa to about 250kDa, about 50kDa to about 200kDa, about 75kDa to about 750kDa, about 75kDa to about 500kDa, about 75 to about 450kDa, about 75 to about 400kDa, about 75kDa to about 350kDa, about 75kDa to about 300kDa, about 75kDa to about 250kDa, about 75kDa to about 200kDa, about 100 to about 100kDa, about 100kDa to about 300kDa, about 100kDa, About 100kDa to about 500kDa, about 100kDa to about 450kDa, about 100kDa to about 400kDa, about 100kDa to about 350kDa, about 100kDa to about 300kDa, about 200kDa to about 750kDa, about 200kDa to about 700kDa, about 200kDa to about 650kDa, about 200kDa to about 600kDa, about 200kDa to about 550kDa, about 200kDa to about 500kDa, about 200kDa to about 450kDa, about 200kDa to about 400kDa, about 250kDa to about 750kDa, about 250kDa to about 700kDa, about 250kDa to about 650kDa, about 250kDa to about 600kDa, about 250kDa to about 550kDa, about 250kDa to about 500kDa, about 250kDa to about 450kDa, about 250kDa to about 400kDa, about 300kDa to about 750, about 300kDa to about 700kDa, about 300kDa to about 650kDa, about 300kDa to about 600kDa, about 300kDa to about 550kDa, or about 300 to about 500 kDa. In a preferred embodiment, the capsular polysaccharide, prior to conjugation, has a molecular weight of from about 25kDa to about 400 kDa. Any integer within any of the above ranges is contemplated as an embodiment of the present disclosure.
In one embodiment of the invention, the serotype Ib capsular polysaccharide comprises its natural sialic acid level, such as about 100% or greater than about 95%. In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (degree of sialylation greater than about 60%), such as up to about 35% (degree of sialylation greater than about 65%), up to about 30% (degree of sialylation greater than about 70%), up to about 25% (degree of sialylation greater than about 75%), up to about 20% (degree of sialylation greater than about 80%), up to about 15% (degree of sialylation greater than about 85%), up to about 10% (degree of sialylation greater than about 90%), or up to about 5% (degree of sialylation greater than about 95%) prior to conjugation.
In another embodiment, serotype Ib capsular polysaccharide has about 1.0mM sialic acid per mM polysaccharide, such as at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation. In another embodiment, the capsular polysaccharide may have at least about 0.6mM sialic acid per mM polysaccharide, such as at least about 0.65mM sialic acid per mM polysaccharide, at least about 0.7mM sialic acid per mM polysaccharide, at least about 0.75mM sialic acid per mM polysaccharide, at least about 0.8mM sialic acid per mM polysaccharide, at least about 0.85mM sialic acid per mM polysaccharide, at least about 0.9mM sialic acid per mM polysaccharide, or at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation.
Serotype Ib capsular polysaccharide is about 0% to about 40% O-acetylated. In one embodiment of the invention, the polysaccharide is de-O-acetylated (i.e., less than about 5% O-acetylated). Some exemplary strains of serotype Ib capsular polysaccharides of the present invention include H36B (ATCC accession number 12401), S40, S42, MB 4053(ATCC accession number 31575), M709, 133, 7357, and PFEGBST 0267.
Serotype II
One embodiment includes a GBS capsular polysaccharide of serotype II. The structure of serotype II can be described as follows:
Figure BDA0003350538080000281
the serotype II capsular polysaccharide, prior to conjugation, has a molecular weight of about 5kDa to about 1,000kDa, such as about 25kDa to about 750kDa, about 25kDa to about 500kDa, about 25kDa to about 450kDa, about 25kDa to about 400kDa, about 25kDa to about 350kDa, about 25kDa to about 300kDa, about 25kDa to about 250kDa, about 25kDa to about 200kDa, about 50kDa to about 750kDa, about 50kDa to about 500kDa, about 50kDa to about 450kDa, about 50kDa to about 400kDa, about 50kDa to about 350kDa, about 50kDa to about 300kDa, about 50kDa to about 250kDa, about 50kDa to about 200kDa, about 75kDa to about 750kDa, about 75kDa to about 500kDa, about 75 to about 450kDa, about 75 to about 400kDa, about 75kDa to about 350kDa, about 75kDa to about 300kDa, about 75kDa to about 250kDa, about 75kDa to about 200kDa, about 100 to about 100kDa, about 100kDa to about 300kDa, about 100kDa, About 100kDa to about 500kDa, about 100kDa to about 450kDa, about 100kDa to about 400kDa, about 100kDa to about 350kDa, about 100kDa to about 300kDa, about 200kDa to about 750kDa, about 200kDa to about 700kDa, about 200kDa to about 650kDa, about 200kDa to about 600kDa, about 200kDa to about 550kDa, about 200kDa to about 500kDa, about 200kDa to about 450kDa, about 200kDa to about 400kDa, about 250kDa to about 750kDa, about 250kDa to about 700kDa, about 250kDa to about 650kDa, about 250kDa to about 600kDa, about 250kDa to about 550kDa, about 250kDa to about 500kDa, about 250kDa to about 450kDa, about 250kDa to about 400kDa, about 300kDa to about 750, about 300kDa to about 700kDa, about 300kDa to about 650kDa, about 300kDa to about 600kDa, about 300kDa to about 550kDa, or about 300 to about 500 kDa. In a preferred embodiment, the capsular polysaccharide, prior to conjugation, has a molecular weight of from about 25kDa to about 400 kDa. Any integer within any of the above ranges is contemplated as an embodiment of the present disclosure.
In one embodiment of the invention, the serotype II capsular polysaccharide comprises its natural sialic acid level, such as about 100% or greater than about 95%. In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (degree of sialylation greater than about 60%), such as up to about 35% (degree of sialylation greater than about 65%), up to about 30% (degree of sialylation greater than about 70%), up to about 25% (degree of sialylation greater than about 75%), up to about 20% (degree of sialylation greater than about 80%), up to about 15% (degree of sialylation greater than about 85%), up to about 10% (degree of sialylation greater than about 90%), or up to about 5% (degree of sialylation greater than about 95%) prior to conjugation.
In another embodiment, serotype II capsular polysaccharide has about 1.0mM sialic acid per mM polysaccharide, such as at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation. In another embodiment, the capsular polysaccharide may have at least about 0.6mM sialic acid per mM polysaccharide, such as at least about 0.65mM sialic acid per mM polysaccharide, at least about 0.7mM sialic acid per mM polysaccharide, at least about 0.75mM sialic acid per mM polysaccharide, at least about 0.8mM sialic acid per mM polysaccharide, at least about 0.85mM sialic acid per mM polysaccharide, at least about 0.9mM sialic acid per mM polysaccharide, or at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation.
The serotype II capsular polysaccharide is less than about 5% O-acetylated. Some exemplary strains of serotype II capsular polysaccharides of the present invention include MB 4055(ATCC accession No. 31576), 18RS21(ATCC accession No. BAA-1175), S16, S20, V8(ATCC accession No. 12973), DK21, DK23, UAB, 5401 and PFEGBST 0708.
Serotype III
One embodiment includes a serotype III GBS capsular polysaccharide. The structure of serotype III can be described as follows:
Figure BDA0003350538080000301
the serotype III capsular polysaccharide, prior to conjugation, has a molecular weight of about 5kDa to about 1,000kDa, such as about 25kDa to about 750kDa, about 25kDa to about 500kDa, about 25kDa to about 450kDa, about 25kDa to about 400kDa, about 25kDa to about 350kDa, about 25kDa to about 300kDa, about 25kDa to about 250kDa, about 25kDa to about 200kDa, about 50kDa to about 750kDa, about 50kDa to about 500kDa, about 50kDa to about 450kDa, about 50kDa to about 400kDa, about 50kDa to about 350kDa, about 50kDa to about 300kDa, about 50kDa to about 250kDa, about 50kDa to about 200kDa, about 75kDa to about 750kDa, about 75kDa to about 500kDa, about 75 to about 450kDa, about 75 to about 400kDa, about 75kDa to about 350kDa, about 75kDa to about 300kDa, about 75kDa to about 250kDa, about 75kDa to about 200kDa, about 100 to about 100kDa, about 100kDa to about 300kDa, about 100kDa, About 100kDa to about 500kDa, about 100kDa to about 450kDa, about 100kDa to about 400kDa, about 100kDa to about 350kDa, about 100kDa to about 300kDa, about 200kDa to about 750kDa, about 200kDa to about 700kDa, about 200kDa to about 650kDa, about 200kDa to about 600kDa, about 200kDa to about 550kDa, about 200kDa to about 500kDa, about 200kDa to about 450kDa, about 200kDa to about 400kDa, about 250kDa to about 750kDa, about 250kDa to about 700kDa, about 250kDa to about 650kDa, about 250kDa to about 600kDa, about 250kDa to about 550kDa, about 250kDa to about 500kDa, about 250kDa to about 450kDa, about 250kDa to about 400kDa, about 300kDa to about 750, about 300kDa to about 700kDa, about 300kDa to about 650kDa, about 300kDa to about 600kDa, about 300kDa to about 550kDa, or about 300 to about 500 kDa. In a preferred embodiment, the capsular polysaccharide, prior to conjugation, has a molecular weight of from about 25kDa to about 200 kDa. In another preferred embodiment, the capsular polysaccharide, prior to conjugation, has a molecular weight of from about 100kDa to about 400 kDa. Any integer within any of the above ranges is contemplated as an embodiment of the present disclosure.
In particular embodiments, the native GBS capsular polysaccharide serotype III is reduced in size using a high pressure homogenization process, but preserves the structural features of the polysaccharide, such as sialic acid.
In one embodiment of the invention, the serotype III capsular polysaccharide comprises its natural sialic acid level, such as about 100% or greater than about 95%. In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (degree of sialylation greater than about 60%), such as up to about 35% (degree of sialylation greater than about 65%), up to about 30% (degree of sialylation greater than about 70%), up to about 25% (degree of sialylation greater than about 75%), up to about 20% (degree of sialylation greater than about 80%), up to about 15% (degree of sialylation greater than about 85%), up to about 10% (degree of sialylation greater than about 90%), or up to about 5% (degree of sialylation greater than about 95%) prior to conjugation.
In another embodiment, serotype III capsular polysaccharide has about 1.0mM sialic acid per mM polysaccharide, such as at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation. In another embodiment, the capsular polysaccharide may have at least about 0.6mM sialic acid per mM polysaccharide, such as at least about 0.65mM sialic acid per mM polysaccharide, at least about 0.7mM sialic acid per mM polysaccharide, at least about 0.75mM sialic acid per mM polysaccharide, at least about 0.8mM sialic acid per mM polysaccharide, at least about 0.85mM sialic acid per mM polysaccharide, at least about 0.9mM sialic acid per mM polysaccharide, or at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation.
Serotype III capsular polysaccharide is about 0% to about 40% O-acetylated. In one embodiment of the invention, the polysaccharide is de-O-acetylated (i.e., O-acetylated less than about 5%). Some exemplary strains of serotype III capsular polysaccharides of the present invention include MB 4082(ATCC accession No. 31577), M132, 110, M781(ATCC accession No. BAA-22), D136C (3) (ATCC accession No. 12403), M782, S23, 120, MB 4316 (M-732; ATCC accession No. 31475), M132, K79, COH1(ATCC accession No. BAA-1176), and PFEGBST 0563.
Serotype IV
One embodiment includes a GBS capsular polysaccharide of serotype IV. The structure of serotype IV can be described as follows:
Figure BDA0003350538080000331
the serotype IV capsular polysaccharide, prior to conjugation, has a molecular weight of about 5kDa to about 1,000kDa, such as about 25kDa to about 750kDa, about 25kDa to about 500kDa, about 25kDa to about 450kDa, about 25kDa to about 400kDa, about 25kDa to about 350kDa, about 25kDa to about 300kDa, about 25kDa to about 250kDa, about 25kDa to about 200kDa, about 50kDa to about 750kDa, about 50kDa to about 500kDa, about 50kDa to about 450kDa, about 50kDa to about 400kDa, about 50kDa to about 350kDa, about 50kDa to about 300kDa, about 50kDa to about 250kDa, about 50kDa to about 200kDa, about 75kDa to about 750kDa, about 75kDa to about 500kDa, about 75 to about 450kDa, about 75 to about 400kDa, about 75kDa to about 350kDa, about 75kDa to about 300kDa, about 75kDa to about 250kDa, about 75kDa to about 200kDa, about 100 to about 100kDa, about 100kDa to about 300kDa, about 100kDa, about 75kDa to about 300kDa, About 100kDa to about 500kDa, about 100kDa to about 450kDa, about 100kDa to about 400kDa, about 100kDa to about 350kDa, about 100kDa to about 300kDa, about 200kDa to about 750kDa, about 200kDa to about 700kDa, about 200kDa to about 650kDa, about 200kDa to about 600kDa, about 200kDa to about 550kDa, about 200kDa to about 500kDa, about 200kDa to about 450kDa, about 200kDa to about 400kDa, about 250kDa to about 750kDa, about 250kDa to about 700kDa, about 250kDa to about 650kDa, about 250kDa to about 600kDa, about 250kDa to about 550kDa, about 250kDa to about 500kDa, about 250kDa to about 450kDa, about 250kDa to about 400kDa, about 300kDa to about 750, about 300kDa to about 700kDa, about 300kDa to about 650kDa, about 300kDa to about 600kDa, about 300kDa to about 550kDa, or about 300 to about 500 kDa. In a preferred embodiment, the capsular polysaccharide, prior to conjugation, has a molecular weight of from about 25kDa to about 400 kDa. Any integer within any of the above ranges is contemplated as an embodiment of the present disclosure.
In one embodiment of the invention, the serotype IV capsular polysaccharide comprises its natural sialic acid level, such as about 100% or greater than about 95%. In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (degree of sialylation greater than about 60%), such as up to about 35% (degree of sialylation greater than about 65%), up to about 30% (degree of sialylation greater than about 70%), up to about 25% (degree of sialylation greater than about 75%), up to about 20% (degree of sialylation greater than about 80%), up to about 15% (degree of sialylation greater than about 85%), up to about 10% (degree of sialylation greater than about 90%), or up to about 5% (degree of sialylation greater than about 95%) prior to conjugation.
In another embodiment, serotype IV capsular polysaccharide has about 1.0mM sialic acid per mM polysaccharide, such as at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation. In another embodiment, the capsular polysaccharide may have at least about 0.6mM sialic acid per mM polysaccharide, such as at least about 0.65mM sialic acid per mM polysaccharide, at least about 0.7mM sialic acid per mM polysaccharide, at least about 0.75mM sialic acid per mM polysaccharide, at least about 0.8mM sialic acid per mM polysaccharide, at least about 0.85mM sialic acid per mM polysaccharide, at least about 0.9mM sialic acid per mM polysaccharide, or at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation.
Serotype IV capsular polysaccharide is about 0% to about 40% O-acetylated. In one embodiment of the invention, the polysaccharide is de-O-acetylated (i.e., O-acetylated less than about 5%). Some exemplary strains of serotype IV capsular polysaccharides of the present invention include 3139(ATCC accession No. 49446), CZ-NI-016 and PFEGBST 0961.
Serotype V
One embodiment includes a serotype V GBS capsular polysaccharide. The structure of serotype V can be described as follows:
Figure BDA0003350538080000351
the serotype V capsular polysaccharide, prior to conjugation, has a molecular weight of about 5kDa to about 1,000kDa, such as about 25kDa to about 750kDa, about 25kDa to about 500kDa, about 25kDa to about 450kDa, about 25kDa to about 400kDa, about 25kDa to about 350kDa, about 25kDa to about 300kDa, about 25kDa to about 250kDa, about 25kDa to about 200kDa, about 50kDa to about 750kDa, about 50kDa to about 500kDa, about 50kDa to about 450kDa, about 50kDa to about 400kDa, about 50kDa to about 350kDa, about 50kDa to about 300kDa, about 50kDa to about 250kDa, about 50kDa to about 200kDa, about 75kDa to about 750kDa, about 75kDa to about 500kDa, about 75 to about 450kDa, about 75 to about 400kDa, about 75kDa to about 350kDa, about 75kDa to about 300kDa, about 75kDa to about 250kDa, about 75kDa to about 200kDa, about 100 to about 100kDa, about 100kDa to about 300kDa, about 100kDa, About 100kDa to about 500kDa, about 100kDa to about 450kDa, about 100kDa to about 400kDa, about 100kDa to about 350kDa, about 100kDa to about 300kDa, about 200kDa to about 750kDa, about 200kDa to about 700kDa, about 200kDa to about 650kDa, about 200kDa to about 600kDa, about 200kDa to about 550kDa, about 200kDa to about 500kDa, about 200kDa to about 450kDa, about 200kDa to about 400kDa, about 250kDa to about 750kDa, about 250kDa to about 700kDa, about 250kDa to about 650kDa, about 250kDa to about 600kDa, about 250kDa to about 550kDa, about 250kDa to about 500kDa, about 250kDa to about 450kDa, about 250kDa to about 400kDa, about 300kDa to about 750, about 300kDa to about 700kDa, about 300kDa to about 650kDa, about 300kDa to about 600kDa, about 300kDa to about 550kDa, or about 300 to about 500 kDa. In a preferred embodiment, the capsular polysaccharide, prior to conjugation, has a molecular weight of from about 25kDa to about 400 kDa. Any integer within any of the above ranges is contemplated as an embodiment of the present disclosure.
In one embodiment of the invention, the serotype V capsular polysaccharide comprises its natural sialic acid level, such as about 100% or greater than about 95%. In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (degree of sialylation greater than about 60%), such as up to about 35% (degree of sialylation greater than about 65%), up to about 30% (degree of sialylation greater than about 70%), up to about 25% (degree of sialylation greater than about 75%), up to about 20% (degree of sialylation greater than about 80%), up to about 15% (degree of sialylation greater than about 85%), up to about 10% (degree of sialylation greater than about 90%), or up to about 5% (degree of sialylation greater than about 95%) prior to conjugation.
In another embodiment, serotype V capsular polysaccharide has about 1.0mM sialic acid per mM polysaccharide, such as at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation. In another embodiment, the capsular polysaccharide may have at least about 0.6mM sialic acid per mM polysaccharide, such as at least about 0.65mM sialic acid per mM polysaccharide, at least about 0.7mM sialic acid per mM polysaccharide, at least about 0.75mM sialic acid per mM polysaccharide, at least about 0.8mM sialic acid per mM polysaccharide, at least about 0.85mM sialic acid per mM polysaccharide, at least about 0.9mM sialic acid per mM polysaccharide, or at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation.
Serotype V capsular polysaccharide is about 0% to about 40% O-acetylated. In one embodiment of the invention, the polysaccharide is de-O-acetylated (i.e., O-acetylated less than about 5%). Some exemplary strains of serotype V capsular polysaccharide of the present invention include 1169-NT1, CJB111(ATCC accession number BAA-23), CJB112, 2603V/R (ATCC accession number BAA-611), NCTC 10/81, CJ11, and PFEGBST 0837.
Serotype VI
GBS serotype VI capsular polysaccharide is described by von Hunolstein, C., et al, Infection and Immunity,6194):1272-1280(1993) (the entire disclosure of which is incorporated herein by reference). The structure of serotype VI can be described as follows:
Figure BDA0003350538080000371
the serotype VI capsular polysaccharide, prior to conjugation, has a molecular weight of about 5kDa to about 1,000kDa, such as about 50kDa to about 750kDa, about 50kDa to about 500kDa, about 50kDa to about 450kDa, about 50kDa to about 400kDa, about 50kDa to about 350kDa, about 50kDa to about 300kDa, about 50kDa to about 250kDa, about 50kDa to about 200kDa, about 75kDa to about 750kDa, about 75kDa to about 500kDa, about 75kDa to about 450kDa, about 75kDa to about 400kDa, about 75kDa to about 350kDa, about 75kDa to about 300kDa, about 75kDa to about 250kDa, about 75kDa to about 200kDa, about 100kDa to about 750kDa, about 100kDa to about 700kDa, about 100kDa to about 650kDa, about 100 to about 600kDa, about 100kDa to about 550kDa, about 100kDa to about 500kDa, about 100kDa to about 450kDa, about 100kDa to about 400kDa, about 100 to about 350kDa, about 100kDa to about 200kDa, about 200kDa to about 200kDa, about 300kDa to about 300kDa, About 200kDa to about 600kDa, about 200kDa to about 550kDa, about 200kDa to about 500kDa, about 200kDa to about 450kDa, about 200kDa to about 400kDa, about 250kDa to about 750kDa, about 250kDa to about 700kDa, about 250kDa to about 650kDa, about 250kDa to about 600kDa, about 250kDa to about 550kDa, about 250kDa to about 500kDa, about 250kDa to about 450kDa, about 250kDa to about 400kDa, about 300kDa to about 750kDa, about 300kDa to about 700kDa, about 300kDa to about 650kDa, about 300kDa to about 600kDa, about 300kDa to about 550kDa, or about 300kDa to about 500 kDa. Any integer within any of the above ranges is contemplated as an embodiment of the present disclosure.
In one embodiment of the invention, the serotype VI capsular polysaccharide comprises its natural sialic acid level, such as about 100% or greater than about 95%. In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (degree of sialylation greater than about 60%), such as up to about 35% (degree of sialylation greater than about 65%), up to about 30% (degree of sialylation greater than about 70%), up to about 25% (degree of sialylation greater than about 75%), up to about 20% (degree of sialylation greater than about 80%), up to about 15% (degree of sialylation greater than about 85%), up to about 10% (degree of sialylation greater than about 90%), or up to about 5% (degree of sialylation greater than about 95%) prior to conjugation.
In another embodiment, serotype VI capsular polysaccharide has about 1.0mM sialic acid per mM polysaccharide, such as at least about 0.95mM sialic acid per mM polysaccharide. In another embodiment, the capsular polysaccharide may have at least about 0.6mM sialic acid per mM polysaccharide, such as at least about 0.65mM sialic acid per mM polysaccharide, at least about 0.7mM sialic acid per mM polysaccharide, at least about 0.75mM sialic acid per mM polysaccharide, at least about 0.8mM sialic acid per mM polysaccharide, at least about 0.85mM sialic acid per mM polysaccharide, at least about 0.9mM sialic acid per mM polysaccharide, or at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation.
Serotype VI capsular polysaccharide is about 0% to about 40% O-acetylated. In one embodiment of the invention, the polysaccharide is de-O-acetylated (i.e., O-acetylated less than about 5%). Some exemplary strains of serotype VI capsular polysaccharide of the invention include 118754, 114852, 114862, 114866, 118775, B4589, B4645, SS1214, and CZ-PW-119.
Serotype VII
GBS serotype VII capsular polysaccharide is described by Kogan, G.et al, Carbohydrate Research,277(1):1-9(1995), the entire disclosure of which is incorporated herein by reference. The repeat units for serotype VII are as follows:
Figure BDA0003350538080000391
the serotype VII capsular polysaccharide, prior to conjugation, has a molecular weight of about 5kDa to about 1,000kDa, such as about 50kDa to about 750kDa, about 50kDa to about 500kDa, about 50kDa to about 450kDa, about 50kDa to about 400kDa, about 50kDa to about 350kDa, about 50kDa to about 300kDa, about 50kDa to about 250kDa, about 50kDa to about 200kDa, about 75kDa to about 750kDa, about 75kDa to about 500kDa, about 75kDa to about 450kDa, about 75kDa to about 400kDa, about 75kDa to about 350kDa, about 75kDa to about 300kDa, about 75kDa to about 250kDa, about 75kDa to about 200kDa, about 100kDa to about 750kDa, about 100kDa to about 700kDa, about 100kDa to about 650kDa, about 100 to about 600kDa, about 100kDa to about 550kDa, about 100kDa to about 500kDa, about 100kDa to about 450kDa, about 100kDa to about 400kDa, about 100 to about 350kDa, about 100kDa to about 200kDa, about 200kDa to about 200kDa, about 300kDa to about 300kDa, About 200kDa to about 600kDa, about 200kDa to about 550kDa, about 200kDa to about 500kDa, about 200kDa to about 450kDa, about 200kDa to about 400kDa, about 250kDa to about 750kDa, about 250kDa to about 700kDa, about 250kDa to about 650kDa, about 250kDa to about 600kDa, about 250kDa to about 550kDa, about 250kDa to about 500kDa, about 250kDa to about 450kDa, about 250kDa to about 400kDa, about 300kDa to about 750kDa, about 300kDa to about 700kDa, about 300kDa to about 650kDa, about 300kDa to about 600kDa, about 300kDa to about 550kDa, or about 300kDa to about 500 kDa. Any integer within any of the above ranges is contemplated as an embodiment of the present disclosure.
In one embodiment of the invention, the serotype VII capsular polysaccharide comprises its natural sialic acid level, such as about 100% or greater than about 95%. In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (degree of sialylation greater than about 60%), such as up to about 35% (degree of sialylation greater than about 65%), up to about 30% (degree of sialylation greater than about 70%), up to about 25% (degree of sialylation greater than about 75%), up to about 20% (degree of sialylation greater than about 80%), up to about 15% (degree of sialylation greater than about 85%), up to about 10% (degree of sialylation greater than about 90%), or up to about 5% (degree of sialylation greater than about 95%) prior to conjugation.
In another embodiment, the serotype VII capsular polysaccharide has about 1.0mM sialic acid per mM polysaccharide, such as at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation. In another embodiment, the capsular polysaccharide may have at least about 0.6mM sialic acid per mM polysaccharide, such as at least about 0.65mM sialic acid per mM polysaccharide, at least about 0.7mM sialic acid per mM polysaccharide, at least about 0.75mM sialic acid per mM polysaccharide, at least about 0.8mM sialic acid per mM polysaccharide, at least about 0.85mM sialic acid per mM polysaccharide, at least about 0.9mM sialic acid per mM polysaccharide, or at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation.
The serotype VII capsular polysaccharide is less than about 5% O-acetylated. Some exemplary strains of serotype VII capsular polysaccharide of the present invention include 7271 and CZ-PW-045.
Serotype VIII
GBS serotype VIII capsular polysaccharide is described by Kogan, G.et al, The Journal of Biological Chemistry,271(15):8786-8790(1996), The entire disclosure of which is incorporated herein by reference. The repeat units for serotype VIII are as follows:
Figure BDA0003350538080000411
the serotype VIII capsular polysaccharide, prior to conjugation, has a molecular weight of about 5kDa to about 1,000kDa, such as about 50kDa to about 750kDa, about 50kDa to about 500kDa, about 50kDa to about 450kDa, about 50kDa to about 400kDa, about 50kDa to about 350kDa, about 50kDa to about 300kDa, about 50kDa to about 250kDa, about 50kDa to about 200kDa, about 75kDa to about 750kDa, about 75kDa to about 500kDa, about 75kDa to about 450kDa, about 75kDa to about 400kDa, about 75kDa to about 350kDa, about 75kDa to about 300kDa, about 75kDa to about 250kDa, about 75kDa to about 200kDa, about 100kDa to about 750kDa, about 100kDa to about 700kDa, about 100kDa to about 650kDa, about 100 to about 600kDa, about 100kDa to about 550kDa, about 100kDa to about 500kDa, about 100kDa to about 450kDa, about 100kDa to about 400kDa, about 100 to about 350kDa, about 100kDa to about 200kDa, about 200kDa to about 200kDa, about 300kDa to about 300kDa, About 200kDa to about 600kDa, about 200kDa to about 550kDa, about 200kDa to about 500kDa, about 200kDa to about 450kDa, about 200kDa to about 400kDa, about 250kDa to about 750kDa, about 250kDa to about 700kDa, about 250kDa to about 650kDa, about 250kDa to about 600kDa, about 250kDa to about 550kDa, about 250kDa to about 500kDa, about 250kDa to about 450kDa, about 250kDa to about 400kDa, about 300kDa to about 750kDa, about 300kDa to about 700kDa, about 300kDa to about 650kDa, about 300kDa to about 600kDa, about 300kDa to about 550kDa, or about 300kDa to about 500 kDa. Any integer within any of the above ranges is contemplated as an embodiment of the present disclosure.
In one embodiment of the invention, the serotype VIII capsular polysaccharide comprises its natural sialic acid level, such as about 100% or greater than about 95%. In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (degree of sialylation greater than about 60%), such as up to about 35% (degree of sialylation greater than about 65%), up to about 30% (degree of sialylation greater than about 70%), up to about 25% (degree of sialylation greater than about 75%), up to about 20% (degree of sialylation greater than about 80%), up to about 15% (degree of sialylation greater than about 85%), up to about 10% (degree of sialylation greater than about 90%), or up to about 5% (degree of sialylation greater than about 95%) prior to conjugation.
In another embodiment, the serotype VIII capsular polysaccharide has about 1.0mM sialic acid per mM polysaccharide, such as at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation. In another embodiment, the capsular polysaccharide may have at least about 0.6mM sialic acid per mM polysaccharide, such as at least about 0.65mM sialic acid per mM polysaccharide, at least about 0.7mM sialic acid per mM polysaccharide, at least about 0.75mM sialic acid per mM polysaccharide, at least about 0.8mM sialic acid per mM polysaccharide, at least about 0.85mM sialic acid per mM polysaccharide, at least about 0.9mM sialic acid per mM polysaccharide, or at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation.
The serotype VIII capsular polysaccharide is about 0% to about 40% O-acetylated. In one embodiment of the invention, the polysaccharide is de-O-acetylated (i.e., O-acetylated less than about 5%). Some exemplary strains of serotype VIII capsular polysaccharides of the present invention include JM9130013 and JM 9130672.
Serotype IX
GBS serotype IX capsular polysaccharide is described by Berti, F., et al, The Journal of Biological Chemistry,289(34):23437-2348(2014), The entire disclosure of which is incorporated herein by reference. The structure of serotype IX can be described as follows:
Figure BDA0003350538080000421
the serotype IX capsular polysaccharide, prior to conjugation, has a molecular weight of about 5kDa to about 1,000kDa, such as about 50kDa to about 750kDa, about 50kDa to about 500kDa, about 50kDa to about 450kDa, about 50kDa to about 400kDa, about 50kDa to about 350kDa, about 50kDa to about 300kDa, about 50kDa to about 250kDa, about 50kDa to about 200kDa, about 75kDa to about 750kDa, about 75kDa to about 500kDa, about 75kDa to about 450kDa, about 75kDa to about 400kDa, about 75kDa to about 350kDa, about 75kDa to about 300kDa, about 75kDa to about 250kDa, about 75kDa to about 200kDa, about 100kDa to about 750kDa, about 100kDa to about 700kDa, about 100kDa to about 650kDa, about 100 to about 600kDa, about 100kDa to about 550kDa, about 100kDa to about 500kDa, about 100kDa to about 450kDa, about 100kDa to about 400kDa, about 100 to about 350kDa, about 100kDa to about 200kDa, about 200kDa to about 200kDa, about 300kDa to about 300kDa, About 200kDa to about 600kDa, about 200kDa to about 550kDa, about 200kDa to about 500kDa, about 200kDa to about 450kDa, about 200kDa to about 400kDa, about 250kDa to about 750kDa, about 250kDa to about 700kDa, about 250kDa to about 650kDa, about 250kDa to about 600kDa, about 250kDa to about 550kDa, about 250kDa to about 500kDa, about 250kDa to about 450kDa, about 250kDa to about 400kDa, about 300kDa to about 750kDa, about 300kDa to about 700kDa, about 300kDa to about 650kDa, about 300kDa to about 600kDa, about 300kDa to about 550kDa, or about 300kDa to about 500 kDa. Any integer within any of the above ranges is contemplated as an embodiment of the present disclosure.
In one embodiment of the invention, the serotype IX capsular polysaccharide comprises its natural sialic acid level, such as about 100% or greater than about 95%. In another embodiment, the capsular polysaccharide may be desialylated up to about 40% (degree of sialylation greater than about 60%), such as up to about 35% (degree of sialylation greater than about 65%), up to about 30% (degree of sialylation greater than about 70%), up to about 25% (degree of sialylation greater than about 75%), up to about 20% (degree of sialylation greater than about 80%), up to about 15% (degree of sialylation greater than about 85%), up to about 10% (degree of sialylation greater than about 90%), or up to about 5% (degree of sialylation greater than about 95%) prior to conjugation.
In another embodiment, serotype IX capsular polysaccharide has about 1.0mM sialic acid per mM polysaccharide, such as at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation. In another embodiment, the capsular polysaccharide may have at least about 0.6mM sialic acid per mM polysaccharide, such as at least about 0.65mM sialic acid per mM polysaccharide, at least about 0.7mM sialic acid per mM polysaccharide, at least about 0.75mM sialic acid per mM polysaccharide, at least about 0.8mM sialic acid per mM polysaccharide, at least about 0.85mM sialic acid per mM polysaccharide, at least about 0.9mM sialic acid per mM polysaccharide, or at least about 0.95mM sialic acid per mM polysaccharide, prior to conjugation.
Serotype IX capsular polysaccharide is about 0% to about 40% O-acetylated. In one embodiment of the invention, the polysaccharide is de-O-acetylated (i.e., O-acetylated less than about 5%). Some exemplary strains of serotype IX capsular polysaccharides of the present invention include IT-NI-016, IT-PW-62, and IT-PW-64.
Polysaccharide-protein conjugates
As used herein, "conjugate" includes a capsular polysaccharide (typically having a desired molecular weight range) and a carrier protein, wherein the capsular polysaccharide is conjugated to the carrier protein. The conjugate may or may not contain some amount of free capsular polysaccharide. As used herein, "free capsular polysaccharide" refers to capsular polysaccharide that is non-covalently associated with (i.e., non-covalently bound to, adsorbed to, or entrapped within) the conjugated capsular polysaccharide-carrier protein. The terms "free capsular polysaccharide", "free polysaccharide" and "free saccharide" are used interchangeably and are intended to convey the same meaning. Regardless of the nature of the carrier molecule, it may be conjugated to the capsular polysaccharide, either directly or through a linker. "conjugation" as used herein refers to the process of covalently linking a bacterial capsular polysaccharide to a carrier molecule. Conjugation enhances the immunogenicity of the bacterial capsular polysaccharide. Conjugation can be performed according to the methods described below or by procedures known in the art.
As used herein, "conjugate immunogenic composition" refers to an immunogenic composition in which the immunogenic agent comprises an antigenic polysaccharide covalently linked to a carrier protein to produce a polysaccharide-protein conjugate. In one embodiment, the polysaccharide-protein conjugates of the invention may be formulated into multivalent immunogenic compositions.
As used herein, the term "molecular weight" of a polysaccharide or carrier protein-polysaccharide conjugate refers to the molecular weight calculated by Size Exclusion Chromatography (SEC) in combination with a multi-angle laser light scattering detector (MALLS).
As used herein, "polysaccharide-protein conjugate" refers to a polysaccharide molecule conjugated to a protein carrier molecule through one or more covalent bonds. It may be desirable to conjugate the polysaccharide to a protein from another species known to be immunogenic in the target host. Thus, in one embodiment, the carrier molecule is a carrier protein. As defined herein, such foreign proteins are referred to as "carrier proteins". The carrier protein is used to enhance the antigenicity and immunogenicity of the polysaccharide. As used herein, the term "carrier effect" refers to the process by which the antigenicity and immunogenicity of a weakly immunogenic or non-immunogenic molecule is enhanced by linking to a more immunogenic molecule (e.g., a heterologous protein) that acts as a carrier. In this case, the polysaccharide in the combined polysaccharide-protein conjugate becomes more immunogenic than when present alone. The carrier protein contains T cell epitopes for stimulation of T cells to facilitate the generation of antibody responses.
As used herein, a "carrier protein" or "proteinA microcarrier "refers to any protein molecule that can be conjugated to an antigen (such as a capsular polysaccharide) to which a desired immune response corresponds. Conjugation of an antigen (such as a polysaccharide) to a carrier protein can render the antigen immunogenic. The carrier protein is preferably a protein that is non-toxic and non-reactogenic and that is available in sufficient quantity and purity. Examples of carrier proteins are toxins, toxoids, or any mutant cross-reactive substances of toxins (CRM) from tetanus, diphtheria, pertussis, Pseudomonas species (Pseudomonas species), Escherichia coli (E.coli), Staphylococcus species (Staphylococcus species), and Streptococcus species (Streptococcus species)197). The carrier protein should be able to withstand standard conjugation procedures. In a particular embodiment of the invention, CRM is used197As a carrier protein.
Cross-reactive materials or CRM are particularly useful for some embodiments of the present invention. Genetically altered proteins can be produced that are antigenically similar to certain bacterial toxins, but are non-toxic. These are called "cross-reactive materials" or CRM. CRM197(Wyeth/Pfizer Inc., Sanford, NC) is notable because it has a single amino acid change from the native diphtheria toxin and is immunologically indistinguishable therefrom. See Pappenheimer, a.m., et al, immunochem, 9(9), 891-906(1972), U.S. Pat. No. 5,614,382, the entire disclosure of which is incorporated herein by reference. CRM 197A non-toxic variant of diphtheria toxin (i.e. toxoid), which was isolated from a culture (culture) of Corynebacterium diphtheriae strain C7(β 197) grown on a medium based on casamino acids and yeast extract. CRM197Purifying by ultrafiltration, ammonium sulfate precipitation and ion exchange chromatography. Manufacture of CRM197A culture of the diphtheria strain C7(C. diphtheria strain C7) (β 197) has been deposited with the american type culture collection (Rockville, Maryland) and has been assigned accession number ATCC 53281. Other diphtheria toxoids are also suitable as carrier proteins. CRM3201 is a genetically engineered variant of pertussis toxin. See Black, W.J., et al, Science,240(4852):656-659(1988), the entire disclosure of which is incorporated herein by reference.
In addition to Diphtheria Toxoid (DT), CRM197And pertussis toxoid, other examples of carrier proteins include Tetanus Toxoid (TT), cholera toxoid (e.g., as described in international patent application publication No. WO 2004/083251), escherichia coli heat-Labile Toxoid (LT), escherichia coli heat-Stable Toxoid (ST), pneumolysin (pneumolysin) from streptococcus pneumoniae (s.pneumona) (wild-type or mutant species with reduced toxicity), pneumococcal surface protein a (pspa), pneumococcal adhesin protein a (psaa), C5a peptidase from streptococcus, hemolysin from staphylococcus aureus, undifferentiated Haemophilus influenzae (nosypaemophilus influenzae) (NTHi) protein, Haemophilus influenzae protein D, Clostridium perfringens (Clostridium perfringens) exotoxin/toxoid, hepatitis B surface antigen, hepatitis B core hepatitis antigen, Rotavirus VP 7 protein and respiratory syncytial virus F and G protein, ovalbumin, Keyhole Limpet Hemocyanin (KLH), Bovine Serum Albumin (BSA), purified tuberculin protein derivative (PPD), and Pseudomonas exotoxin or its derivative, including recombinantly produced non-toxic mutant Pseudomonas aeruginosa exotoxin A. Bacterial outer membrane proteins such as outer membrane protein mixture c (ompc), porin (porin), transferrin binding protein, or clostridium difficile enterotoxin (toxin a) and cytotoxin (toxin B) may also be used. Other proteins, such as ovalbumin, Keyhole Limpet Hemocyanin (KLH), Bovine Serum Albumin (BSA) or purified protein derivatives of tuberculin (PPD) may also be used as carrier proteins. In a preferred embodiment, the carrier protein is diphtheria toxoid. More preferably, the carrier protein is CRM 197. In another embodiment of the invention, the carrier protein is tetanus toxoid.
To synthesize multivalent conjugate immunogenic compositions, polysaccharide-protein conjugates can be produced by conjugating a mixture of polysaccharides purified from two different bacterial species to a carrier protein. Alternatively, multivalent conjugate immunogenic compositions can be made by combining polysaccharides purified from two or more different serotypes of the same bacterium and conjugating them in mixture with a carrier protein. Alternatively, polysaccharide-protein conjugates produced by reacting a single type of polysaccharide with a carrier protein in separate reactions (using different polysaccharides) can be mixed. Thus, multivalent immunogenic compositions may include a carrier protein with a homogeneous or heterogeneous population of linked polysaccharides.
After conjugation of the capsular polysaccharide to the carrier protein, the polysaccharide-protein conjugate (enriched in terms of the amount of polysaccharide-protein conjugate) is purified by various techniques. These include, for example, concentration/diafiltration operations, precipitation/elution, column chromatography and depth filtration.
As described above, the present invention relates to conjugates comprising a GBS capsular polysaccharide conjugated to a carrier protein. An embodiment of the invention provides a conjugate comprising a GBS serotype IV capsular polysaccharide conjugated to a carrier protein and at least one additional conjugate comprising a GBS serotype la capsular polysaccharide conjugated to a carrier protein, a GBS serotype lb capsular polysaccharide conjugated to a carrier protein, a GBS serotype II capsular polysaccharide conjugated to a carrier protein, a GBS serotype III capsular polysaccharide conjugated to a carrier protein, a GBS serotype V capsular polysaccharide conjugated to a carrier protein, a GBS serotype VI capsular polysaccharide conjugated to a carrier protein, a GBS serotype VII capsular polysaccharide conjugated to a carrier protein, a GBS serotype VIII capsular polysaccharide conjugated to a carrier protein, or a GBS serotype IX capsular polysaccharide conjugated to a carrier protein. In one aspect of the invention, the polysaccharide has a molecular weight of about 5kDa to 1,000 kDa; the molecular weight of the conjugate is from about 300kDa to about 20,000 kDa; and the conjugate comprises less than about 40% free polysaccharide compared to the total amount of polysaccharide. In one embodiment, the conjugate comprises less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, or less than about 5% free polysaccharide compared to the total amount of polysaccharide.
In one embodiment, the serotype Ia, Ib, II, III, IV, V, VI, VII, VIII, and/or IX polysaccharide has a molecular weight of about 5kDa to about 1,000kDa, such as about 50kDa to about 750kDa, about 50kDa to about 500kDa, about 50kDa to about 450kDa, about 50kDa to about 400kDa, about 50kDa to about 350kDa, about 50kDa to about 300kDa, about 50kDa to about 250kDa, about 50kDa to about 200kDa, about 75kDa to about 750kDa, about 75kDa to about 500kDa, about 75kDa to about 450kDa, about 75kDa to about 400kDa, about 75kDa to about 350kDa, about 75kDa to about 300kDa, about 75kDa to about 250kDa, about 75 to about 200kDa, about 100 to about 750kDa, about 100kDa to about 700kDa, about 100kDa to about 650kDa, about 100kDa to about 600kDa, about 100 to about 550kDa, about 100 to about 100kDa to about 500kDa, about 100kDa to about 300kDa, about 100kDa to about 100kDa, From about 200kDa to about 750kDa, from about 200kDa to about 700kDa, from about 200kDa to about 650kDa, from about 200kDa to about 600kDa, from about 200kDa to about 550kDa, from about 200kDa to about 500kDa, from about 200kDa to about 450kDa, from about 200kDa to about 400kDa, from about 250kDa to about 750kDa, from about 250kDa to about 700kDa, from about 250kDa to about 650kDa, from about 250kDa to about 600kDa, from about 250kDa to about 550kDa, from about 250kDa to about 500kDa, from about 250kDa to about 450kDa, from about 250kDa to about 400kDa, from about 300kDa to about 750kDa, from about 300kDa to about 700kDa, from about 300kDa to about 650kDa, from about 300kDa to about 600kDa, from about 300kDa to about 550kDa, or from about 300kDa to about 500 kDa. Any integer within any of the above ranges is contemplated as an embodiment of the present disclosure.
In one embodiment, the conjugate has a molecular weight of about 300kDa to about 20,000kDa, such as about 300kDa to about 15,000kDa, about 300kDa to about 10,000kDa, about 300kDa to about 9,000kDa, about 300kDa to about 8,000kDa, about 300kDa to about 7,000kDa, about 300kDa to about 6,000kDa, about 300kDa to about 5,000kDa, about 300kDa to about 4,000kDa, about 300kDa to about 3,000kDa, about 300kDa to about 2,000kDa, about 300kDa to about 1,000kDa, about 500kDa to about 20,000kDa, about 500kDa to about 15,000kDa, about 500kDa to about 10,000kDa, about 500kDa to about 9,000kDa, about 500 to about 8,000kDa, about 500kDa to about 7,000kDa, about 500kDa to about 6,000kDa, about 500kDa to about 5,000kDa, about 500 to about 4000kDa, about 3 kDa to about 8,000kDa, about 1,000kDa to about 1,000kDa, about 500kDa to about 1,000kDa, about 1,000kDa to about 1,000kDa, about 500kDa to about 1,000kDa, about 1,000kDa to about 1,000, about 1,000kDa to about 1,000kDa, about 10,000, About 1,000kDa to about 6,000kDa, about 1,000kDa to about 5,000kDa, about 1,500kDa to about 20,000kDa, about 1,500kDa to about 15,000kDa, about 1,500kDa to about 10,000kDa, about 1,500kDa to about 9,000kDa, about 1,500kDa to about 8,000kDa, about 1,500kDa to about 7,000kDa, about 1,500kDa to about 6,000kDa, about 1500kDa to about 5,000kDa, about 2,000kDa to about 20,000kDa, about 2,000kDa to about 15,000kDa, about 2,000kDa to about 10,000kDa, about 2,000kDa to about 9,000kDa, about 2,000kDa to about 8,000kDa, about 2,000kDa to about 7,000kDa, about 2,000kDa to about 6,000kDa, about 2,500kDa to about 20,000kDa, about 2,000kDa to about 10,000kDa, about 3,000kDa to about 3,000kDa, about 3,000kDa to about 3,000 kDa.
In one embodiment, the GBS serotype IV capsular polysaccharide conjugate has a molecular weight in any of the ranges described above.
In one embodiment, the GBS serotype Ia capsular polysaccharide conjugate has a molecular weight in any of the ranges described above.
In one embodiment, the GBS serotype Ib capsular polysaccharide conjugate has a molecular weight in any of the ranges described above.
In one embodiment, the GBS serotype II capsular polysaccharide conjugate has a molecular weight in any of the ranges described above.
In one embodiment, the GBS serotype III capsular polysaccharide conjugate has a molecular weight in any of the ranges described above.
In one embodiment, the GBS serotype V capsular polysaccharide conjugate has a molecular weight in any of the ranges described above.
In one embodiment, the GBS serotype VI capsular polysaccharide conjugate has a molecular weight in any of the ranges described above.
In one embodiment, the GBS serotype VII capsular polysaccharide conjugate has a molecular weight in any of the ranges described above.
In one embodiment, the GBS serotype VIII capsular polysaccharide conjugate has a molecular weight in any of the ranges described above.
In one embodiment, the GBS serotype IX capsular polysaccharide conjugate has a molecular weight in any of the ranges described above.
In one embodiment, a conjugate of the invention has at least about 0.6, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9, 0.95, 0.97, or 0.98mM sialic acid per mM polysaccharide. In a preferred embodiment, the conjugate has at least about 0.9 or 0.95mM sialic acid per mM polysaccharide.
In one embodiment, the GBS serotype IV capsular polysaccharide conjugate has a sialic acid content of at least any of the above values.
In one embodiment, the GBS serotype Ia capsular polysaccharide conjugate has a sialic acid content of at least any one of the above values.
In one embodiment, the GBS serotype Ib capsular polysaccharide conjugate has a sialic acid content of at least any one of the above values.
In one embodiment, the GBS serotype II capsular polysaccharide conjugate has a sialic acid content of at least any of the above values.
In one embodiment, the GBS serotype III capsular polysaccharide conjugate has a sialic acid content of at least any of the above values.
In one embodiment, the GBS serotype V capsular polysaccharide conjugate has a sialic acid content of at least any of the above values.
In one embodiment, the GBS serotype VI capsular polysaccharide conjugate has a sialic acid content of at least any of the above values.
In one embodiment, the GBS serotype VII capsular polysaccharide conjugate has a sialic acid content of at least any of the above values.
In one embodiment, the GBS serotype VIII capsular polysaccharide conjugate has a sialic acid content of at least any one of the above values.
In one embodiment, the GBS serotype IX capsular polysaccharide conjugate has a sialic acid content of at least any of the above values.
In one embodiment, the conjugates of the invention have less than about 0.01, 0.02, 0.03, 0.04, or 0.05mM O-acetate (O-acetate) per mM saccharide repeat unit. In another embodiment, the conjugate comprises at least about 0.1, 0.2, 0.3, 0.35, or about 0.4mM O-acetate per mM saccharide repeat unit.
In one embodiment, the GBS serotype IV capsular polysaccharide conjugate has an O-acetate content of any of the values described above.
In one embodiment, the GBS serotype Ia capsular polysaccharide conjugate has an O-acetate content of any of the values described above.
In one embodiment, the GBS serotype Ib capsular polysaccharide conjugate has an O-acetate content of any of the values described above.
In one embodiment, the GBS serotype II capsular polysaccharide conjugate has an O-acetate content of any of the values described above.
In one embodiment, the GBS serotype III capsular polysaccharide conjugate has an O-acetate content of any of the values described above.
In one embodiment, the GBS serotype V capsular polysaccharide conjugate has an O-acetate content of any of the values described above.
In one embodiment, the GBS serotype VI capsular polysaccharide conjugate has an O-acetate content of any of the values described above.
In one embodiment, the GBS serotype VII capsular polysaccharide conjugate has an O-acetate content of any of the values described above.
In one embodiment, the GBS serotype VIII capsular polysaccharide conjugate has an O-acetate content of any of the values described above.
In one embodiment, the GBS serotype IX capsular polysaccharide conjugate has an O-acetate content of any of the values described above.
In another embodiment, the immunogenic conjugate comprises less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, or less than about 5% free GBS capsular polysaccharide compared to the total amount of GBS capsular polysaccharide. In a preferred embodiment, the immunogenic conjugate comprises less than about 5% unreacted free saccharide compared to the total amount of GBS capsular polysaccharide.
In another embodiment, the ratio (weight to weight) of GBS capsular polysaccharide to carrier protein in the conjugate is from about 0.5 to about 3.0. In one aspect, the ratio of GBS capsular polysaccharide to carrier protein in the conjugate is from about 0.5 to about 2.0, from about 0.5 to about 1.5, from about 0.5 to about 1.0, from about 1.0 to about 1.5, or from about 1.0 to about 2.0. In a preferred embodiment, the ratio of GBS capsular polysaccharide to carrier protein in the conjugate is from about 0.8 to about 1.0.
In another embodiment, the degree of conjugation of the conjugate is 2 to 15, 2 to 13, 2 to 10, 2 to 8, 2 to 6, 2 to 5, 2 to 4, 3 to 15, 3 to 13, 3 to 10, 3 to 8, 3 to 6, 3 to 5, 3 to 4, 5 to 15, 5 to 10, 8 to 15, 8 to 12, 10 to 15, or 10 to 12. In a preferred embodiment, the degree of conjugation of the conjugate is from 2 to 5.
Conjugation
Conjugation may be direct, in which atoms from the polysaccharide are covalently bonded to atoms from the protein surface. Alternatively, conjugation may be through a linker molecule that reacts with both the polysaccharide and the protein and links the two, thereby tethering the carbohydrate to the protein.
When the carrier is conjugated (i.e., covalently bound) to one or more antigens, such as polysaccharides, the conjugation can be carried out by any chemical method, process, or genetic technique known in the art. For example, the carrier polypeptide and one or more antigens selected from the group comprising carbohydrates, oligosaccharides, lipids, lipooligosaccharides, polysaccharides, oligosaccharide-protein conjugates, polysaccharide-protein conjugates, peptide-protein conjugates, oligosaccharide-peptide conjugates, polysaccharide-peptide conjugates, protein-protein conjugates, lipooligosaccharide-protein conjugates, polysaccharide-protein conjugates, or any combination thereof, can be conjugated by a variety of techniques, including, but not limited to: (1) direct coupling via a protein functional group (e.g., thiol-thiol linkage, amine-carboxyl linkage, amine-aldehyde linkage; enzyme direct coupling); (2) homobifunctional coupling of amines (e.g., using bis-aldehydes); (3) homobifunctional coupling of thiols (e.g., using bis-maleimides); (4) homobifunctional coupling by photoactivated reagents; (5) heterobifunctional coupling of amines to thiols (e.g., using maleimides); (6) heterobifunctional coupling by photoactivated reagents (e.g., β -carbonyl diazo group); (7) introducing amine reactive groups into the polysaccharide or oligosaccharide by cyanogen bromide activation or carboxymethylation; (8) introduction of thiol-reactive groups into the polysaccharide or oligosaccharide by heterobifunctional compounds (such as maleimido-hydrazide); (9) protein-lipid conjugation by introducing hydrophobic groups into proteins and (10) protein-lipid conjugation by incorporating reactive groups into lipids. In addition, heterobifunctional "non-covalent coupling" techniques (such as biotin-avidin interaction) are under consideration. Other methods for conjugating oligo-and polysaccharides to immunogenic carrier proteins well known in the art are also within the scope of certain embodiments of the invention.
In one embodiment, the GBS capsular polysaccharide-protein conjugate is obtained by activating the polysaccharide with 1-cyano-4-dimethylaminopyridinium tetrafluoroborate (CDAP) to form a cyanate ester. The activated polysaccharide may be coupled to the amino groups on the carrier protein directly or via a spacer (linker). For example, the spacer can be cystamine or cysteamine to produce a thiolated polysaccharide that can be coupled to a support via a thioether linkage obtained after reaction with a maleimide-activated carrier protein (e.g., using GMBS) or a haloacetylated carrier protein (e.g., using iodoacetamide, SIB, SIAB, sulfo-SIAB, SIA, or SBAP).
In one aspect, the cyanate ester (optionally made by CDAP chemistry) is coupled to hexanediamine or Adipic Dihydrazide (ADH), while the amino-derivatized saccharide is conjugated to the carrier protein through a carboxyl group on the protein carrier using carbodiimide (e.g., EDAC or EDC) chemistry. Such conjugates are described, for example, in international patent application publication nos. WO 93/15760, WO 95/08348 and WO 96/29094.
Other suitable techniques use carbodiimides, hydrazides, active esters, norbornane, p-nitrobenzoic acid, N-hydroxysuccinimide, S- -NHS, EDC and TSTU. International patent application publication No. WO 98/42721 has many descriptions. Conjugation may involve a carbonyl linker which may be formed by reacting the free hydroxyl group of the saccharide with 1, 1-Carbonyldiimidazole (CDI) or 1, 1-carbonylbis-1, 2, 4-triazole (CDT) (see Bethell, et al, J.biol. chem.,254: 2572-. This may involve reducing the mutarotameric terminus to a primary hydroxyl group, optionally protecting/deprotecting the primary hydroxyl group, reacting the primary hydroxyl group with CDI/CDT to form a CDI/CDT carbamate intermediate, and coupling the CDI/CDT carbamate intermediate to an amino group on the protein.
In a preferred embodiment, the GBS capsular polysaccharide-protein conjugate of the invention is manufactured using reductive amination. Reductive amination involves two steps: (1) oxidizing the polysaccharide to produce aldehyde functions from adjacent diols in the individual hexose units and (2) reducing the activated polysaccharide and the carrier protein to form a conjugate.
In one embodiment, the GBS capsular polysaccharide is activated (oxidized) by a process comprising the steps of:
(a) reacting the isolated GBS capsular polysaccharide with an oxidizing agent; and
(b) the oxidation reaction is quenched by addition of a quencher to produce an activated GBS capsular polysaccharide.
In one aspect of the invention, the concentration of the isolated capsular polysaccharide is from about 0.1mg/mL to about 10.0mg/mL, such as from about 0.5mg/mL to about 5.0mg/mL, from about 1.0mg/mL to about 3.0mg/mL, or about 2.0 mg/mL.
In a particular embodiment, the oxidizing agent is periodate. Periodate oxidizes adjacent hydroxyl groups to form carbonyl or aldehyde groups and causes C-C bond cleavage. The term "periodate" includes both periodate and periodic acid. The term also includes the partial periodate (IO) salt4 -) And ortho-periodate (IO)6 5-). The term "periodate" also includes various periodates, including sodium periodate and potassium periodate. In a preferred embodiment, the oxidizing agent is sodium periodate. In a preferred embodiment, the periodate used to oxidize the GBS capsular polysaccharide is a meta-periodate. In a preferred embodiment, the periodate salt used to oxidize the serotype capsular polysaccharide is sodium metaperiodate.
In another embodiment, the polysaccharide is reacted with 0.01 to 10.0, 0.05 to 5.0, 0.1 to 1.0, 0.5 to 1.0, 0.7 to 0.8, 0.05 to 0.5, or 0.1 to 0.3 molar equivalents of an oxidizing agent. In particular embodiments, the polysaccharide is reacted with about 0.05, about 0.1, about 0.15, about 0.2, about 0.25, about 0.3, about 0.35, about 0.4, about 0.45, about 0.5, about 0.55, about 0.6, about 0.65, about 0.7, about 0.75, about 0.8, about 0.85, about 0.9, or about 0.95 molar equivalents of oxidizing agent. In another embodiment, the polysaccharide is reacted with about 0.1 molar equivalents of an oxidizing agent. In another embodiment, the polysaccharide is reacted with about 0.15 molar equivalents of an oxidizing agent. In another embodiment, the polysaccharide is reacted with about 0.25 molar equivalents of an oxidizing agent. In another embodiment, the polysaccharide is reacted with about 0.5 molar equivalents of an oxidizing agent. In an alternative embodiment, the polysaccharide is reacted with about 0.6 molar equivalents of oxidizing agent. In another embodiment, the polysaccharide is reacted with about 0.7 molar equivalents of an oxidizing agent.
In one aspect of the invention, the duration of the oxidation reaction is from about 1 hour to about 50 hours, from about 10 hours to about 30 hours, from about 15 hours to about 20 hours, from about 15 hours to about 17 hours, or about 16 hours.
In another aspect of the invention, the temperature of the oxidation reaction is maintained at about 2 ℃ to about 25 ℃, about 2 ℃ to about 8 ℃, or about 20 ℃ to about 25 ℃. In a preferred embodiment, the temperature of the reaction is maintained at about 23 ℃. In another preferred embodiment, the temperature of the reaction is maintained at about 5 ℃.
In another aspect, the oxidation reaction is carried out in a buffer selected from the group consisting of: sodium phosphate, potassium phosphate, 2- (N-morpholino) ethanesulfonic acid (MES) and Bis-Tris. In a preferred embodiment, the buffering agent is potassium phosphate.
In further aspects, the buffer is at a concentration of about 1mM to about 500mM, about 1mM to about 300mM, or about 50mM to about 200 mM. In a preferred embodiment, the buffer is present at a concentration of about 100 mM.
In one aspect, the oxidation reaction is carried out at a pH of about 4.0 to about 8.0, about 5.0 to about 7.0, or about 5.5 to about 6.5. In a preferred embodiment, the pH is about 6.0.
In one embodiment, the activated GBS capsular polysaccharide is obtained by reacting about 0.5mg/L to about 5.0mg/mL of the isolated capsular polysaccharide with about 0.05 to about 0.3 molar equivalents of periodate at a temperature of about 20 ℃ to 25 ℃.
In another embodiment, the activated GBS capsular polysaccharide is obtained by reacting about 0.5mg/L to about 5.0mg/mL of the isolated capsular polysaccharide with about 0.05 to about 0.3 molar equivalents of periodate at a temperature of about 2 ℃ to 8 ℃.
In another embodiment, the activated GBS capsular polysaccharide is purified according to methods known to those skilled in the art, such as Gel Permeation Chromatography (GPC), dialysis, or ultrafiltration/diafiltration. For example, the activated capsular polysaccharide is purified by concentration and diafiltration using an ultrafiltration device.
In one embodiment, the activated GBS capsular polysaccharide has a degree of oxidation of 5 to 25, for example 5 to 15, 5 to 10, 10 to 25, 10 to 20, 10 to 15. In a preferred embodiment, the activated GBS capsular polysaccharide has a degree of oxidation of 10 to 20, 11 to 19, 12 to 18, 13 to 17, or 14 to 16.
In another embodiment, the activated GBS capsular polysaccharide has a molecular weight of from about 5kDa to about 1,000kDa, such as from about 50kDa to about 300kDa, from about 75kDa to about 400kDa, from about 75kDa to about 200kDa, from about 100kDa to about 700kDa, from about 100kDa to about 500kDa, from about 100kDa to about 400kDa, from about 100kDa to about 300kDa, from about 200kDa to about 400kDa, from about 300kDa to about 700 kDa. In a preferred embodiment, the activated GBS capsular polysaccharide has a molecular weight of from about 75kDa to about 400 kDa.
In one embodiment, the activated GBS capsular polysaccharide is freeze-dried, optionally in the presence of a saccharide. In a preferred embodiment, the sugar is selected from sucrose, trehalose (trehalose), raffinose, stachyose (stachyose), melezitose (melezitose), dextran (dextran), mannitol, lactitol (lactitol) and palatinit (palatinit). In a preferred embodiment, the sugar is sucrose. The freeze-dried activated capsular polysaccharide may then be mixed with a solution comprising a carrier protein.
In another embodiment, the activated GBS capsular polysaccharide is mixed with a carrier protein, optionally in the presence of a saccharide, and freeze-dried. In one aspect, the sugar is selected from sucrose, trehalose, raffinose, stachyose, melezitose, dextran, mannitol, lactitol and xylitol. In a preferred embodiment, the sugar is sucrose. The co-lyophilized polysaccharide and carrier protein may then be resuspended in solution and reacted with a reducing agent.
The activated GBS capsular polysaccharide may be conjugated to a carrier protein by a process comprising the steps of:
(a) mixing the activated GBS capsular polysaccharide with a carrier protein,
(b) reacting the mixed activated GBS capsular polysaccharide and carrier protein with a reducing agent to form a GBS capsular polysaccharide-carrier protein conjugate.
For example, conjugation of activated GBS capsular polysaccharides to carrier proteins by reductive amination in polar aprotic solvents is suitable to maintain low levels of free polysaccharide, compared to reductive amination in aqueous solution, where the amount of unreacted (free) polysaccharide is significantly elevated. In a preferred embodiment, step (a) and step (b) are carried out in a polar aprotic solvent.
In one embodiment, step (a) comprises dissolving the freeze-dried GBS capsular polysaccharide in a solution comprising a carrier protein and a polar aprotic solvent. In another embodiment, step (a) comprises dissolving the co-lyophilized GBS capsular polysaccharide and carrier protein in a polar aprotic solvent.
In one embodiment, the polar aprotic solvent is selected from the group consisting of: dimethyl sulfoxide (DMSO), sulfolane, Dimethylformamide (DMF), and Hexamethylphosphoramide (HMPA). In a preferred embodiment, the polar aprotic solvent is DMSO.
When steps (a) and (b) are performed in an aqueous solution, steps (a) and (b) are performed in a buffer, preferably selected from PBS, MES, HEPES, Bis-tris, ADA, PIPES, MOPSO, BES, MOPS, DIPSO, MOBS, HEPPSO, POPSO, TEA, EPPS, Bicine, or HEPB, in an aqueous medium at a pH of about 6.0 to about 8.5, about 7.0 to about 8.0, or about 7.0 to about 7.5. In a preferred embodiment, the buffer is PBS. In a preferred embodiment, the pH is about 7.3.
In one embodiment, the concentration of the activated GBS capsular polysaccharide in step (b) is from about 0.1mg/mL to about 10.0mg/mL, from about 0.5mg/mL to about 5.0mg/mL, or from about 0.5mg/mL to about 2.0 mg/mL. In a preferred embodiment, the activated serotype GBS capsular polysaccharide in step (b) is at a concentration of about 0.1mg/mL, about 0.2mg/mL, about 0.3mg/mL, about 0.4mg/mL, about 0.5mg/mL, about 0.6mg/mL, about 0.7mg/mL, about 0.8mg/mL, about 0.9mg/mL, about 1.0mg/mL, about 1.1mg/mL, about 1.2mg/mL, about 1.3mg/mL, about 1.4mg/mL, about 1.5mg/mL, about 1.6mg/mL, about 1.7mg/mL, about 1.8mg/mL, about 1.9mg/mL, about 2.0mg/mL, about 2.1mg/mL, about 2.2mg/mL, about 2.3mg/mL, about 2.4mg/mL, about 2.5mg/mL, about 2.6mg/mL, about 2.0mg/mL, about 2.1mg/mL, about 2.2mg/mL, about 2.3mg/mL, about 2.4mg/mL, about 2.5mg/mL, about 2.6mg/mL, about 2mg/mL, about 2.6mg/mL, about 2mg/mL, about 2.6mg/mL, or, About 2.9mg/mL, or about 3.0 mg/mL.
In a preferred embodiment, the initial ratio (weight to weight) of activated GBS capsular polysaccharide of serotype to carrier protein is 5: 1 to 0.1: 1. 2: 1 to 0.1: 1. 2: 1 to 1: 1. 1.5: 1 to 1: 1. 0.1: 1 to 1: 1. 0.3: 1 to 1: 1. 0.6: 1 to 1: 1. in a preferred embodiment, the initial ratio of activated GBS capsular polysaccharide of serotype to carrier protein is about 0.4: 1. 0.5: 1. 0.6: 1. 0.7: 1. 0.8: 1. 0.9: 1. 1: 1. 1.1: 1. 1.2: 1. 1.3: 1. 1.4: 1. 1.5: 1. 1.6: 1. 1.7: 1. 1.8: 1. 1.9: 1. 2: 1.
in one embodiment, the reducing agent is sodium cyanoborohydride, sodium triacetoxyborohydride, sodium borohydride and zinc borohydride in the presence of a Bronsted or Lewis acid, an amine borane such as pyridine borane, 2-picoline borane, 2, 6-diborane-methanol, dimethylamine-borane, t-BuMeiPrN-BH3benzylamine-BH3Or 5-ethyl-2-picoline borane (PEMB). In a preferred embodiment, the reducing agent is sodium cyanoborohydride.
In another embodiment, the amount of reducing agent used in step (b) is from about 0.1 to about 10.0 molar equivalents, from about 0.5 to about 5.0 molar equivalents, or from about 1.0 to about 2.0 molar equivalents. In a preferred embodiment, the amount of reducing agent used in step (b) is about 1.0, 1.1, 1.2, 1.3, 1.4, 1.5, 1.6, 1.7, 1.8, 1.9, or 2.0 molar equivalents.
In a preferred embodiment, the duration of step (b) is from 1 hour to 60 hours, from 10 hours to 50 hours, from 40 hours to 50 hours, or from 42 hours to 46 hours. In a preferred embodiment, the duration of step (b) is about 44 hours.
In another embodiment, the reaction temperature of step (b) is maintained at 10 ℃ to 40 ℃, 15 ℃ to 30 ℃, or 20 ℃ to 26 ℃. In a preferred embodiment, the reaction temperature in step (b) is maintained at about 23 ℃.
In further embodiments, the method for manufacturing an immunogenic conjugate comprising a GBS capsular polysaccharide covalently linked to a carrier protein further comprises the step of capping (quenching) unreacted aldehyde by adding borohydride (step (c)).
In one embodiment, the capping agent is a borohydride selected from the group consisting of: sodium borohydride (NaBH)4) Sodium cyanoborohydride, lithium borohydride, potassium borohydride, tetrabutylammonium borohydride, calcium borohydride and magnesium borohydride. In a preferred embodiment, the capping reagent is sodium borohydride.
In another embodiment, the amount of borohydride used in step (c) is from about 0.1 to about 10.0 molar equivalents, from about 0.5 to about 5.0 molar equivalents, or from about 1.0 to about 3.0 molar equivalents. In a preferred embodiment, the amount of borohydride used in step (c) is about 2.0 molar equivalents.
In a preferred embodiment, the borohydride used in step (c) is NaBH4The concentration is about 2.0 molar equivalents.
In one embodiment, the duration of step (c) is from 0.1 hour to 10 hours, from 0.5 hour to 5 hours, from 2 hours to 4 hours. In a preferred embodiment, the duration of step (c) is about 3 hours.
In another embodiment, the reaction temperature in step (c) is maintained at about 15 ℃ to about 45 ℃, about 15 ℃ to about 30 ℃, or about 20 ℃ to about 26 ℃. In a preferred embodiment, the reaction temperature of step (c) is maintained at about 23 ℃.
After conjugation and capping of the GBS capsular polysaccharide to the carrier protein, the polysaccharide-protein conjugate can be purified (enriched in terms of the amount of polysaccharide-protein conjugate) by various techniques known to those skilled in the art. These include dialysis, concentration/diafiltration operations, tangential flow filtration, precipitation/elution, column chromatography (DEAE or hydrophobic interaction chromatography), and depth filtration.
In another embodiment, the immunogenic conjugate comprises less than about 40%, less than about 35%, less than about 30%, less than about 25%, less than about 20%, less than about 15%, less than about 10%, or less than about 5% free GBS capsular polysaccharide as compared to the total amount of GBS capsular polysaccharide. In a preferred embodiment, the immunogenic conjugate comprises less than about 5% unreacted free saccharide compared to the total amount of GBS capsular polysaccharide.
In a preferred embodiment, the GBS polysaccharide-protein conjugate has a molecular weight of about 300kDa to about 20,000kDa, such as about 1,000kDa to about 15,000kDa, or about 1,000kDa to about 10,000 kDa.
In another embodiment, the ratio (weight/weight) of GBS capsular polysaccharide to carrier protein in the conjugate is from about 0.5 to about 3.0. In one aspect, the ratio of GBS capsular polysaccharide to carrier protein in the conjugate is from about 0.5 to about 2.0, from about 0.5 to about 1.5, from about 0.5 to about 1.0, from about 1.0 to about 1.5, or from about 1.0 to about 2.0. In a preferred embodiment, the ratio of GBS capsular polysaccharide to carrier protein in the conjugate is from about 0.8 to about 1.0.
In another embodiment, the degree of conjugation of the conjugate is 2 to 15, 2 to 13, 2 to 10, 2 to 8, 2 to 6, 2 to 5, 2 to 4, 3 to 15, 3 to 13, 3 to 10, 3 to 8, 3 to 6, 3 to 5, 3 to 4, 5 to 15, 5 to 10, 8 to 15, 8 to 12, 10 to 15, or 10 to 12. In a preferred embodiment, the degree of conjugation of the conjugate is from 2 to 5.
In one aspect of the invention, the GBS capsular polysaccharide-protein conjugate is obtained by the reductive amination process described above. For example, in one aspect, the present disclosure provides GBS capsular polysaccharide-protein conjugates comprising a polysaccharide conjugated to a carrier protein, which may be made or obtained by a process comprising the steps of:
(a) Reacting the isolated GBS capsular polysaccharide with an oxidizing agent;
(b) quenching the oxidation reaction by adding a quencher to produce an activated GBS capsular polysaccharide;
(c) mixing the activated GBS capsular polysaccharide with a carrier protein,
(d) reacting the mixed activated GBS capsular polysaccharide and carrier protein with a reducing agent to form a GBS capsular polysaccharide-carrier protein conjugate, and, optionally
(e) By adding sodium borohydride (NaBH)4) To cap the unreacted aldehyde.
In a preferred embodiment, steps (c) and (d) are carried out in DMSO.
In another aspect of the invention, the GBS capsular polysaccharide-protein conjugate of the invention is made using reductive amination as described above, but using 2,2,6, 6-tetramethyl-1-piperidinyloxy (TEMPO) radical and N-chlorosuccinimide (NCS) as co-oxidants in the activation/oxidation step. See international patent application publication No. WO 2014/097099, the entire contents of which are incorporated herein by reference. In such embodiments, glycoconjugates from GBS capsular polysaccharides are made using TEMPO free radical to oxidize primary alcohols of saccharides (primary alcohols) to aldehydes (using NCS as co-oxidant) (hereinafter "TEMPO/NCS oxidation"), such as described in example 7 and international patent application publication No. WO 2014/097099. Thus, in one aspect, a conjugate of a GBS capsular polysaccharide is obtained by a process comprising the steps of: a) reacting GBS capsular polysaccharide with TEMPO and NCS in a solvent to produce an activated saccharide; and b) reacting the activated saccharide with a carrier protein comprising one or more amino groups (hereinafter referred to as "TEMPO/NCS-reductive amination"). In one embodiment, the solvent may be an aqueous solvent or DMSO.
In one aspect, the GBS capsular polysaccharide-protein conjugate is obtained by the method. For example, in one aspect, the present disclosure provides a GBS capsular polysaccharide-protein conjugate comprising a polysaccharide conjugated to a carrier protein made or obtained by a process comprising the steps of: a) reacting a saccharide with 2,2,6, 6-tetramethyl-1-piperidinyloxy (TEMPO) and N-chlorosuccinimide (NCS) in a solvent to produce an activated saccharide; and b) reacting the activated saccharide with a carrier protein comprising one or more amino groups. In one embodiment, the solvent may be an aqueous solvent or DMSO.
Immunogenic compositions
After purification of the individual conjugates, they can be combined to formulate immunogenic compositions of the invention, which can be used, for example, in vaccines. The formulation of the immunogenic compositions of the invention can be accomplished using art-recognized methods.
An "immune response" to an immunogenic composition is a subject's development of a humoral and/or cell-mediated immune response to a molecule (e.g., an antigen, such as a protein or polysaccharide) present in the composition of interest. For the purposes of the present invention, a "humoral immune response" is an immune response mediated by antibodies and involves the production of antibodies with affinity for the antigens present in the immunogenic composition of the invention, whereas a "cell-mediated immune response" is an immune response mediated by T-lymphocytes and/or other leukocytes. A "cell-mediated immune response" is caused by the presentation of antigenic epitopes associated with class I or class II molecules of the Major Histocompatibility Complex (MHC). This activates antigen-specific CD4+ T helper cells or CD8+ Cytotoxic T Lymphocytes (CTLs). CTLs are specific for peptide or lipid antigens presented in association with MHC or proteins encoded by CD1 and expressed on the cell surface. CTLs help to induce and promote intracellular destruction of intracellular microorganisms or lysis of cells infected by such microorganisms. Another aspect of cellular immunity relates to antigen-specific responses produced by helper T cells. Helper T cells act on and focus on the function of, and activity of, assisting in the stimulation of non-specific effector cells against cells displaying peptide antigens on their surface (associated with classical or non-classical MHC molecules). "cell-mediated immune response" also refers to the production of cytokines, chemokines, and other such molecules made by activated T cells and/or other leukocytes, including those derived from CD4+ and CD8+ T cells. The ability of a particular antigen or composition to stimulate a cell-mediated immune response can be determined by a variety of assays, such as by lymphoproliferation (lymphocyte activation) assays, CTL cytotoxic cell assays, by assaying for T lymphocytes specific for the antigen in a sensitized subject, or by measuring the production of cytokines by T cells in response to antigen restimulation. Such assays are well known in the art. See, e.g., Erickson, A.L., et al, J.Immunol, 151(8):4189-4199 (1993); doe, B.et al, Eur.J.Immunol.24(10): 2369-.
The term "immunogenicity" refers to the ability of an antigen or vaccine to elicit an immune response (humoral or cell-mediated, or both).
As used herein, "immunogenic amount," or "immunologically effective amount," or "dose," each of which are used interchangeably, generally refers to the amount of antigen or immunogenic composition (as measured by standard assays known to those skilled in the art) that is sufficient to elicit an immune response that is a cellular (T cell), or humoral (B cell or antibody) response, or both.
As used herein, "immune interference" or "significant immune interference" refers to a statistically significant reduction in an immune response to an individual antigen in a multivalent or multicomponent vaccine as compared to an immune response to the same antigen administered in a monovalent vaccine.
A "protective" immune response refers to the ability of an immunogenic composition to elicit an immune response (humoral or cell-mediated) that is used to protect a subject from infection. The protection provided need not be absolute, i.e., need not completely prevent or eradicate infection, if there is a statistically significant improvement when compared to a population of control subjects (e.g., infected animals not administered a vaccine or immunogenic composition). Protection may be limited to reducing the severity or rate of onset of symptoms of the infection. Several assays are known in the art for determining whether an immune response appears to be a "protective immune response". For example, increased antibody levels can be measured by a binding assay, such as a whole cell ELISA assay described further below. Other assays include measuring functional antibody responses, such as promoting killing of bacteria, which can be tested with an opsonophagocytosis assay (OPA) as described below. In particular instances, a "protective immune response" may comprise inducing a two-fold or four-fold increase in antibody levels specific for a particular antigen in at least 50% of subjects. In another instance, a "protective immune response" can include a reduction in bacterial number of at least 10%, 25%, 50%, 65%, 75%, 80%, 85%, 90%, 95%, or more.
The amount of a particular conjugate in a composition is typically calculated based on the total polysaccharide (conjugated and unconjugated) used for the conjugate. For example, in a GBS capsular polysaccharide dose of 100mcg/ml, a GBS capsular polysaccharide conjugate with 20% free polysaccharide will have about 80mcg/ml conjugated GBS capsular polysaccharide and about 20mcg/ml unconjugated GBS capsular polysaccharide. The contribution of the protein carrier to the conjugate is generally not taken into account when calculating the dose of the conjugate. The amount of conjugate may vary depending on the streptococcus serotype. Typically, each dose will comprise from about 0.01mg/ml to about 100mcg/ml of each polysaccharide, especially from about 1mcg/ml to about 70mcg/ml, more especially from about 5mcg/ml to about 50 mcg/ml. The "immunogenic amount" of the different polysaccharide components in the immunogenic composition can be different and can each include about 0.01mcg/ml, about 0.1mcg/ml, about 0.25mcg/ml, about 0.5mcg/ml, about 1mcg/ml, about 2mcg/ml, about 3mcg/ml, about 4mcg/ml, about 5mcg/ml, about 6mcg/ml, about 7mcg/ml, about 8mcg/ml, about 9mcg/ml, about 10mcg/ml, about 15mcg/ml, about 20mcg/ml, about 25mcg/ml, about 30mcg/ml, about 40mcg/ml, about 50mcg/ml, about 60mcg/ml, about 70mcg/ml, about 80mcg/ml, about 90mcg/ml, about 100mcg/ml of any particular polysaccharide antigen. Unless otherwise indicated, the dosage or immunogenic amount of a multivalent immunogenic composition will refer to the dosage of each polysaccharide. For example, a 10mcg/ml dose of hexavalent immunogenic composition will contain 10mcg/ml of each of the six polysaccharides.
The efficacy of an antigen as an immunogen can be determined by measuring the level of B cell activity (by measuring the level of circulating antibodies specific for the antigen in serum using an immunoassay, an immunoprecipitation assay, a functional antibody assay such as an in vitro opsonin assay, and many other assays known in the art). Another method of measuring the efficacy of an antigen as a T cell immunogen can be determined by a proliferation assay, by a cell lysis assay (such as a chromium release assay) to measure the ability of a T cell to lyse its specific target cell. In addition, in the present invention, an "immunogenic amount" can also be defined by measuring the serum level of antigen-specific antibodies induced following administration of the antigen, or by measuring the ability of the antibodies induced thereby to enhance the opsonophagocytic capacity of particular leukocytes as described herein. The level of protection of the immune response can be measured using the injected antigen to challenge the immunized host. For example, if the antigen required for an immune response is a bacterium, the level of protection induced by the "immunogenic amount" of that antigen can be measured by measuring the percent survival or percent death following challenge of the animal with bacterial cells. In one embodiment, the protective amount can be determined by measuring at least one symptom associated with the bacterial infection (e.g., fever associated with the infection). The amount of each antigen in a multiple antigen or multicomponent vaccine or immunogenic composition will vary with respect to the other components and can be determined by methods known to those skilled in the art. Such methods may include, for example, procedures for measuring immunogenicity and/or in vivo efficacy.
The term "immunogenic composition" relates to any pharmaceutical composition containing an antigen (e.g., a microorganism or a component thereof) that can be used to elicit an immune response in a subject. The immunogenic compositions of the invention may be used to treat a human susceptible to GBS infection by administering the immunogenic composition via a systemic transdermal or mucosal route. These administrations may include injection via intramuscular (i.m.), intraperitoneal (i.p.), intradermal (i.d.), or subcutaneous routes; administration via a patch or other transdermal delivery device; or by mucosal administration to the oral/digestive, respiratory or genitourinary tract. In one embodiment, the immunogenic composition can be used in the manufacture of a vaccine or for eliciting polyclonal or monoclonal antibodies that can be used for passive protection or treatment of animals.
In one aspect, the invention relates to an immunogenic composition comprising an effective amount of at least one polysaccharide, oligosaccharide, polysaccharide-protein conjugate, or bioequivalent thereof as described herein. For example, in one embodiment, the immunogenic composition comprises a polysaccharide-protein conjugate, wherein the capsular polysaccharide is selected from the group consisting of: group B streptococcal serotypes Ia, Ib, II, III, IV, V, VI, VII, VIII and IX, and wherein the capsular polysaccharide has a sialic acid level of greater than about 60%. In another example, an immunogenic composition comprises a polysaccharide-protein conjugate, wherein the conjugate comprises a capsular polysaccharide from group B streptococcus serotype IV and at least one additional serotype selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In another embodiment, the immunogenic composition comprises a polysaccharide-protein conjugate, wherein the conjugate comprises a capsular polysaccharide from group B streptococcus serotype IV and at least two additional serotypes selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In another embodiment, the immunogenic composition comprises a polysaccharide-protein conjugate, wherein the conjugate comprises a capsular polysaccharide from group B streptococcus serotype IV and at least three additional serotypes selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In another embodiment, the immunogenic composition comprises a polysaccharide-protein conjugate. Wherein the conjugate comprises capsular polysaccharides from group B Streptococcus serotype IV and at least four additional serotypes selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In a particular embodiment, the immunogenic composition comprises a polysaccharide-protein conjugate, wherein the conjugate comprises capsular polysaccharides from group B streptococcus serotypes Ia, Ib, II, III and V. In another embodiment, the immunogenic composition comprises a polysaccharide-protein conjugate, wherein the conjugate comprises capsular polysaccharides from group B streptococcus serotypes Ia, Ib, II, III, and IV. In another embodiment, the immunogenic composition comprises a polysaccharide-protein conjugate, wherein the conjugate comprises capsular polysaccharides from group B streptococcus serotype IV and at least five additional serotypes selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In one such embodiment, the immunogenic composition comprises six polysaccharide-protein conjugates, wherein the conjugates comprise capsular polysaccharides from group B streptococcus serotypes Ia, Ib, II, III, IV, and V.
In one embodiment, the immunogenic composition of the invention comprises 2 to 10 different serotypes of streptococcus agalactiae. Thus, in one embodiment, the immunogenic composition of the invention is a GBS conjugate composition having a valency of 2, 3, 4, 5, 6, 7, 8, 9 or 10. In one such embodiment, the immunogenic composition is a 5-valent GBS conjugate composition. In another embodiment, the immunogenic composition is a 6-valent GBS conjugate composition. In another embodiment, the immunogenic composition is a 7-valent GBS conjugate composition. In another embodiment, the immunogenic composition is an 8-valent GBS conjugate composition.
Although the prior art teaches the use of less than six, less than five, or less than four GBS antigens in a composition (see international patent application publication nos. WO 2006/082527 and WO 2006/082530) and experiences immune interference, particularly with respect to the use of serotype V in a multivalent composition (see international patent application publication No. WO 2012/035519), the present invention does not show any significant immune interference when using four or more GBS antigens in a multivalent composition and serotype V. Accordingly, the present invention relates to a multivalent immunogenic composition comprising a polysaccharide-protein conjugate comprising at least four GBS capsular polysaccharide serotypes, such as at least five GBS capsular polysaccharide serotypes, at least six GBS capsular polysaccharide serotypes, at least seven GBS capsular polysaccharide serotypes, at least eight GBS capsular polysaccharide serotypes, or at least nine GBS capsular polysaccharide serotypes, wherein the composition does not have significant immunological interference. In a particular embodiment, the immunogenic composition comprises GBS capsular polysaccharide serotype V.
The polysaccharide-protein conjugate may comprise the same or different protein carriers. In one embodiment, the conjugate comprises the same protein carrier and the saccharide is conjugated to the same molecule of the protein carrier (the carrier molecule has 2 or more different polysaccharides conjugated to it) [ see, e.g., international patent application publication No. WO 2004/083251 ]. In another embodiment, the polysaccharides are each independently conjugated to a different molecule of the protein carrier (each molecule of the protein carrier having only one type of polysaccharide conjugated thereto). In this embodiment, it is described that capsular saccharides are individually conjugated to carrier proteins.
The optimal component amounts for a particular immunogenic composition can be determined by standard studies involving observation of the appropriate immune response in a subject. Following initial vaccination, the subject may receive one or more appropriately spaced boosters.
In addition to the plurality of capsular polysaccharide protein conjugates, the immunogenic compositions of the invention may further comprise one or more preservatives. The FDA requires that the bioproduct in multi-dose vials contain preservatives with few exceptions. The present invention contemplates the use of such multi-dose vials. Preservative containing vaccine products include vaccines containing benzethonium chloride (anthrax), 2-phenoxyethanol (DTaP, HepA, Lyme, Polio (Polio) (parenteral) and phenol (Pneumo, typhoid (parenteral)). Preservatives approved for use in injectable pharmaceuticals include, for example, chlorobutanol, m-cresol, methyl paraben, propyl paraben, 2-phenoxyethanol, benxonine chloride, benzyldimethylammonium chloride, benzyl acid, benzyl alcohol, phenol, and phenylmercuric nitrate.
In another aspect, the invention relates to a composition comprising at least one polysaccharide of any described herein and a pharmaceutically acceptable excipient, buffer, stabilizer, adjuvant, cryoprotectant, salt, divalent cation, non-ionic detergent, free radical oxidation inhibitor, diluent or carrier, or mixture thereof.
The immunogenic composition optionally may include one or more physiologically acceptable buffers selected from, but not limited to, HEPES, PIPES, MES, Tris (tromethamine), phosphate, acetate, borate, citrate, glycine, histidine and succinate. In a preferred embodiment, the buffering agent is histidine.
In one embodiment, the immunogenic composition comprises a buffer at a concentration of about 5mM to about 50mM, about 5mM to about 40mM, about 5mM to about 30mM, about 5mM to about 20mM, about 5mM to about 10mM, about 10mM to about 50mM, about 10mM to about 40mM, about 10mM to about 35mM, about 10mM to about 30mM, about 10mM to about 25mM, about 10mM to about 20mM, about 10mM to about 15mM, about 15mM to about 50mM, about 15mM to about 40mM, about 15mM to about 35mM, about 15mM to about 30mM, about 15mM to about 25mM, or about 15mM to about 20 mM. In a preferred embodiment, the immunogenic composition comprises a buffer at a concentration of about 10mM to about 25mM, and most preferably about 20 mM.
In a preferred embodiment, the immunogenic composition comprises histidine at a concentration of about 20 mM.
In some embodiments, the formulation is buffered to a pH in the range of about 5.0 to about 7.1, such as about 5.3 to about 7.1, about 5.5 to about 7.0, about 6.0 to about 6.5, about 6.3 to about 7.0, or about 6.5 to about 7.0. In another embodiment, the formulation is buffered to a pH of about 6.0, 6.1, 6.2, 6.3, 6.4, 6.5, 6.6, 6.7, 6.8, 6.9, or 7.0. In a preferred embodiment, the formulation is buffered to a pH in the range of about 6.0 to about 7.0, and most preferably about 6.5.
The immunogenic composition optionally may comprise one or more nonionic surfactants including, but not limited to, polyoxyethylene sorbitan fatty acid esters, polysorbate-80 (TWEEN 80), polysorbate-60 (TWEEN 60), polysorbate-40 (TWEEN 40), polysorbate-20 (TWEEN 20), and polyoxyethylene alkyl ethers (polyoxyethylene alkyl ethers), including, but not limited to BRIJ 58, BRIJ 35, and others such as TRITON X-100; TRITON X-114, NP40, SPAN 85 and PLURONIC series of nonionic surfactants (e.g. PLURONIC 121). In one embodiment, the immunogenic composition comprises polysorbate-80 or polysorbate 40, preferably polysorbate-80 (PS 80).
In one embodiment, the immunogenic composition comprises a surfactant at a concentration of about 0.001% to about 2% (v/w), about 0.001% to about 1%, about 0.001% to about 0.5%, about 0.001% to about 0.1%, about 0.001% to about 0.05%, about 0.001% to about 0.01%, about 0.001% to 0.005%, about 0.005% to about 2%, about 0.005% to about 1%, about 0.005% to about 0.5%, about 0.005% to about 0.1%, about 0.005% to about 0.05%, about 0.005% to about 0.01%, about 0.01% to about 2%, about 0.01% to about 1%, about 0.01% to about 0.5%, about 0.01% to about 0.1%, about 0.01% to about 0.05%, about 0.01% to about 0.015%, about 0.015% to about 0.015%, about 0.03% to about 0.015%, about 0.05% to about 0.05%, about 0.001% to about 0.05%, about 0.015% to about 0.01% to about 0.015%, about 0.1%, about 0.015% to about 0.1%, about 0.015%, about 0.1%, about 0.015% to about 0.1%, about 0.015%, about 0.1% to about 0.1%, about 0.015%, about 0.05%, about 0., About 0.02% to about 1%, about 0.02% to about 0.5%, about 0.02% to about 0.1%, about 0.02% to about 0.05%, about 0.02% to about 0.04%, about 0.02% to about 0.03%, about 0.05% to about 2%, about 0.05% to about 1%, about 0.05% to about 0.5%, about 0.05% to about 0.1%, about 0.1% to about 2%, about 0.1% to about 1%, about 0.1% to about 0.5%, or about 0.1% to about 0.25%. In a preferred embodiment, the immunogenic composition comprises a surfactant at a concentration of about 0.01% to about 0.03%, and most preferably about 0.02%.
In another embodiment, the immunogenic composition comprises polysorbate-80 at a concentration of about 0.001% to about 2% (preferably up to about 0.25%) or polysorbate 40 at a concentration of about 0.001% to 1% (preferably up to about 0.5%).
In a preferred embodiment, the immunogenic composition comprises PS80 at a concentration of about 0.02%.
Pharmaceutically acceptable carriers are not to be confused with "carrier proteins" which are used to link the carbohydrates of the invention to proteins and to modify the immune response to the carbohydrates. To avoid confusion with the carrier proteins described herein, the term pharmaceutically acceptable diluent will be preferred over a pharmaceutically acceptable carrier, although these terms may occasionally be used interchangeably. The term "pharmaceutically acceptable carrier" means a carrier that is approved by a regulatory agency of the federal or a state government or other regulatory agency, or a carrier that is listed in the U.S. pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans and non-human mammals. The term "carrier" refers to a diluent, adjuvant, excipient, or vehicle with which the pharmaceutical composition is administered. Suitable pharmaceutically acceptable diluents include any and all conventional solvents, dispersion media, fillers, solid carriers, aqueous solutions, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like. Such pharmaceutically acceptable diluents can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin. Water, water for injection (WFI), sterile isotonic saline solution, phosphate buffered saline, adjuvant suspensions, aqueous dextrose and glycerol solutions, and combinations thereof, can be employed as the liquid carrier, particularly for injectable solutions. Pharmaceutically acceptable diluents may further include minor amounts of auxiliary substances such as wetting or emulsifying agents, preservatives or buffering agents which may enhance shelf life or efficacy in the body. The preparation and use of pharmaceutically acceptable diluents are known in the art. Examples of suitable Pharmaceutical carriers are described in "Remington's Pharmaceutical Sciences" of e.w. martin. In one embodiment, the diluent is water, water for injection (WFI), adjuvant suspension, or saline. In particular embodiments, the diluent is a suspension of any of the adjuvants described herein. In a preferred embodiment, the diluent is an aluminum-based adjuvant suspension, such as an aluminum phosphate suspension.
Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, trehalose, raffinose, stachyose, melezitose, dextran, mannitol, lactitol, xylitol, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, glycine, arginine, lysine, sodium chloride (NaCl), dried skim milk, glycerol, propylene glycol, water, ethanol and the like. In a preferred embodiment, the excipient is NaCl.
In one embodiment, the immunogenic composition comprises an excipient at a concentration of about 10mM to about 500mM, about 10mM to about 450mM, about 10mM to about 400mM, about 10mM to about 350mM, about 10mM to about 300mM, about 10mM to about 250mM, about 10mM to about 200mM, about 10mM to about 150mM, about 10mM to about 100mM, about 10mM to about 50mM, about 10mM to about 30mM, about 10mM to about 20mM, 20mM to about 500mM, about 20mM to about 450mM, about 20mM to about 400mM, about 20mM to about 350mM, about 20mM to about 300mM, about 20mM to about 250mM, about 20mM to about 200mM, about 20mM to about 150mM, about 20mM to about 100mM, about 20mM to about 50mM, about 20mM to about 30mM, 50mM to about 500mM, about 50mM to about 450mM, about 50mM to about 50mM, about 50mM to about 400mM, about 50mM to about 50mM, about 50mM to about 300mM, about 50mM, about 300mM, about 250mM, about 50mM, about 300mM, about 50mM, about 250mM, about 50mM, about 300mM, about 250mM, about 50mM, about 250mM, about 200mM, about 250mM, about 50mM, about 200mM, about 250mM, about 50mM, about 200mM, about 50mM, about 250mM, about 50mM, about 200mM, about 50mM, about 250mM, about 200mM, about 50mM, about 250mM, about 50mM, about 250mM, about 50mM, about 200mM, about 250mM, about 50mM, about 250mM, about 50mM, about 200mM, about 50mM, about 250mM, about 50mM, about 250mM, about 50, About 50mM to about 150mM, about 50mM to about 100mM, about 100mM to about 500mM, about 100mM to about 450mM, about 100mM to about 400mM, about 100mM to about 350mM, about 100mM to about 300mM, about 100mM to about 250mM, about 100mM to about 200mM, about 100mM to about 150mM, about 150mM to about 500mM, about 150mM to about 450mM, about 150mM to about 400mM, about 150mM to about 350mM, about 150mM to about 300mM, about 150mM to about 250mM, about 150mM to about 200mM, about 200mM to about 500mM, about 200mM to about 450mM, about 200mM to about 400mM, about 200mM to about 350mM, about 200mM to about 300mM, about 200mM to about 250mM, about 250mM to about 500mM, about 250mM to about 450mM, about 250mM to about 400mM, about 250mM to about 350mM, about 300mM to about 300mM, about 250mM to about 300mM, about 300mM to about 300mM, about 500mM to about 500mM, about 250mM, about 500mM, about 150mM, about 500mM, about 150mM, about 500mM, about, About 350mM to about 450mM, about 350mM to about 400mM, about 400mM to about 500mM, about 400mM to about 450mM, or about 450mM to about 500 mM. In a preferred embodiment, the immunogenic composition comprises an excipient at a concentration of about 10mM to about 250mM, most preferably about 150 mM.
In a preferred embodiment, the excipient is NaCl at a concentration of about 150 mM.
If desired, the compositions may also contain minor amounts of wetting agents, bulking agents, emulsifying agents, or pH buffering agents. These compositions may be in the form of solutions, suspensions, emulsions, freeze-dried powders or blocks, and the like. The formulation should be suitable for the mode of administration. Any conventional media or agent, except those incompatible with the active ingredient, is contemplated for use in the immunogenic compositions of the invention.
In one embodiment, the immunogenic composition is freeze-dried (optionally in the presence of at least one excipient). In a preferred embodiment, the at least one excipient is selected from the group consisting of: starch, glucose, lactose, sucrose, trehalose, raffinose, stachyose, melezitose, dextran, mannitol, lactitol, arabitol, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, glycine, arginine, lysine, sodium chloride (NaCl), dried skim milk, glycerol, propylene glycol, water and ethanol. In a preferred embodiment, the at least one excipient is selected from the group consisting of: sucrose, mannitol, and glycine. In a particular embodiment, the at least one excipient is sucrose. In another embodiment, the freeze-dried composition comprises additional excipients. In one such embodiment, the additional excipient is mannitol or glycine.
In another embodiment, the freeze-dried composition comprises from about 1% (w/v) to about 10% (w/v) of at least one sugar, such as about 1.5%, 2.0%, 2.5%, 3.0%, 3.5%, 4.0%, 4.5%, 5.0%, 5.5%, 6.0%, 6.5%, 7.0%, 7.5%, 8.0%, 8.5%, 9.0%, 9.5%, or 10.0%. In a preferred embodiment, the freeze-dried composition comprises more than about 5.5% (w/v) of at least one excipient, such as more than about 7.0% (w/v). In another embodiment, the lyophilized composition comprises from about 1% (w/v) to about 10% (w/v) of an additional excipient, such as about 1.5%, 2.0%, 2.5%, 3.0%, 3.5%, 4.0%, 4.5%, 5.0%, 5.5%, 6.0%, 6.5%, 7.0%, 7.5%, 8.0%, 8.5%, 9.0%, 9.5%, or 10.0%. In a preferred embodiment, the lyophilized composition comprises from about 1% (w/v) to about 10% (w/v) of at least one excipient and from about 1% (w/v) to about 10% (w/v) of an additional excipient.
In another embodiment, the freeze-dried composition is reconstituted with water, water for injection (WFI), adjuvant suspension, or saline. In a preferred embodiment, the diluent is an aluminum-based adjuvant suspension, such as an aluminum phosphate suspension.
In one embodiment, the composition comprises an isolated polysaccharide described herein and a carrier molecule. Suitable carrier molecules may include proteins, polysaccharides, polylactic acid, polyglycolic acid, polymeric amino acids, amino acid copolymers, lipid aggregates (such as oil droplets or liposomes), and inactive viral particles. Examples of particulate carriers include those derived from polymethylmethacrylate polymers, as well as microparticles derived from poly (lactide) and poly (lactide-co-glycolide) (referred to as PLG).
The immunogenic compositions of the invention may further comprise one or more additional "immunomodulators", which are agents that disrupt or alter the immune system such that up-or down-regulation of humoral and/or cell-mediated immunity is observed. In particular embodiments, it is preferred to upregulate humoral and/or cell-mediated branches of the immune system (arm). Examples of some immunomodulatory agents include, for example, adjuvants or cytokines as described in U.S. patent No. 5,254,339, or ISCOMATRIX (CSL Limited, Parkville, Australia). The term "adjuvant" refers to a compound or mixture that enhances an immune response to an antigen as further described herein.
Non-limiting examples of adjuvants that can be used in the compositions of the present invention include RIBI adjuvant system (RIBI inc., Hamilton, Mont.); mineral gels, such as aluminum hydroxide gel; water-in-oil emulsions, such as freund's complete and incomplete adjuvants; block copolymers (CytRx, Atlanta Ga.); SAF-M (Chiron, Emeryville, Calif.);
Figure BDA0003350538080000711
an adjuvant; a saponin; quil a or other saponin fractions; monophosphoryl lipid a; and an alfidrine (Avridine) lipid-amine adjuvant. Non-limiting examples of oil-in-water emulsions that can be used as adjuvants in the vaccines of the present invention include MF59 (U.S. Pat. No. 6,299,884) (containing 5% Squalene (Squalene), 0.5% polysorbate-80, and 0.5% Span 85 (optionally containing various amounts of MTP-PE) formulated as submicron particles using a microfluidizer such as model 110Y microfluidizer (Microfluidics, Newton, MA)), and SAF (containing 10% Squalene, 0.4% polysorbate-80, 5% pluronic block polymer L121, and thr-MDP microfluidized into submicron emulsions or shaken into submicron emulsionsMixing to produce a larger particle size emulsion); modified SEAM62 (containing 5% (v/v) squalene (Sigma), 1% (v/v)
Figure BDA0003350538080000712
Detergents (ICI Surfactants), 0.7% (v/v) polysorbate 80 detergent (ICI Surfactants), 2.5% (v/v) ethanol, 200. mu.g/ml Quil A, 100. mu.g/ml cholesterol, and 0.5% (v/v) lecithin; and modified SEAM 1/2 (containing 5% (v/v) squalene, 1% (v/v)
Figure BDA0003350538080000713
Detergent, 0.7% (v/v) polysorbate 80 detergent, 2.5% (v/v) ethanol, 100 μ g/ml Quil a, and 50 μ g/ml cholesterol).
Suitable adjuvants for enhancing an immune response also include, but are not limited to, MPL as described in U.S. Pat. No. 4,912,094TM(3-O-deacylated monophosphoryl lipid A, Corixa, Hamilton, MT). Also suitable as adjuvants are synthetic lipid a analogs, or amine alkyl glucosamine phosphate compounds (AGPs), or derivatives or analogs thereof, available from Corixa (Hamilton, MT) and described in U.S. patent No. 6,113,918. One such AGP is 2- [ (R) -3-tetradecanoyloxy-tetradecanoyl-amino]Ethyl 2-deoxy-4-O-phosphono-3-O- [ (R) -3-tetradecanoyloxy-tetradecanoyl]-2- [ (R) -3-tetradecanoyloxy-tetradecanoyl-amino]-b-D-glucopyranoside, which is also known as 529 (formerly RC 529). This 529 adjuvant is formulated as an Aqueous Form (AF) or as a Stable Emulsion (SE).
Other adjuvants include cyclodextrin derivatives (U.S. Pat. No. 6,165,995); polyanionic polymers (polyanical polymers) (U.S. Pat. No. 6,610,310); muramyl peptides (muramyl peptides), such as N-acetyl-muramyl-L-threoaminoyl-D-isoglutamine (thr-MDP) and N-acetyl-orthopoly-L-alanine-2- (1'-2' dipalmitoyl-sn-glycero-3-hydroxy-phosphoryl-oxy) -ethylamine (MTP-PE); aifenugine (Amphigen); avridine (Avridine); l121/squalene; d-lactide-polylactic acid/glycoside; a pluronic polyol; killed Bordetella (killed Bordetella); saponins, such as saponins of U.S. patent No. 5, 057,540 StimulonTMQS-21 (antibiotics, Framingham, MA.); mycobacterium tuberculosis; bacterial lipopolysaccharides; synthetic polynucleotides, such as oligonucleotides containing CpG motifs (e.g., U.S. Pat. No. 6,207,646); IC-31(Intercell AG, Vienna, Austria) as described in European patent Nos. 1,296,713 and 1,326,634; pertussis Toxin (PT) or a mutant thereof, cholera toxin or a mutant thereof (e.g., U.S. Pat. nos. 7,285,281, 7,332,174, 7,361,355, and 7,384,640); or E.coli heat-Labile Toxin (LT) or mutants thereof, especially LT-K63, LT-R72 (e.g., U.S. Pat. Nos. 6,149,919, 7,115,730 and 7,291,588).
Other "immunomodulators" which may be included in a vaccine include, for example, one or more of the following: interleukins 1-alpha, 1-beta, 2, 4, 5, 6,7, 8, 10, 12 (see, e.g., U.S. Pat. No. 5,723,127), 13, 14, 15, 16, 17 and 18 (and mutant forms thereof); interferon- α, β and γ; granulocyte-macrophage colony stimulating factor (GM-CSF) (see, e.g., U.S. Pat. No. 5,078,996 and ATCC accession No. 39900); macrophage colony stimulating factor (M-CSF); granulocyte colony stimulating factor (G-CSF); or tumor necrosis factors alpha and beta. Still other adjuvants that may be used in the immunogenic compositions described herein include chemokines, including, but not limited to, MCP-1, MIP-1 α, MIP-1 β, and RANTES; adhesion molecules, such as selectins, e.g., L-selectin, P-selectin and E-selectin; mucin-like molecules such as CD34, GlyCAM-1 and MadCAM-1; members of the integrin family, such as LFA-1, VLA-1, Mac-1, and p 150.95; members of the immunoglobulin superfamily, such as PECAM, ICAMs (e.g., ICAM-1, ICAM-2, and ICAM-3), CD2, and LFA-3; co-stimulatory molecules such as B7-1, B7-2, CD40, and CD 40L; growth factors including vascular growth factor, nerve growth factor, fibroblast growth factor, epidermal growth factor, PDGF, BL-1 and vascular endothelial growth factor; receptor molecules including Fas, TNF receptor, Flt, Apo-1, p55, WSL-1, DR3, TRAMP, Apo-3, AIR, LARD, NGRF, DR4, DR5, KILLER, TRAIL-R2, TRICK2, and DR 6; and Caspase (ICE).
It will be understood that the decision whether or not to use an immunomodulator and/or adjuvant, or the choice of which immunomodulator and/or adjuvant to use, will depend on the subject to which the vaccine or immunogenic composition is to be administered, the route of injection and the number of injections to be given. For example, if the subject has been naturally exposed to a pathogen, an adjuvant may not be required, as the vaccine antigen can effectively elicit a memory response. In some embodiments, the immunogenic composition will include one or more adjuvants. In one embodiment, the immunogenic composition comprises an aluminum-based adjuvant. In one such embodiment, the aluminum adjuvant is aluminum hydroxide, aluminum phosphate, or aluminum hydroxyphosphate. In a particular embodiment, the adjuvant is aluminum phosphate. In another embodiment of the invention, the immunogenic composition comprises QS-21 as an adjuvant.
In one embodiment, the immunogenic composition comprises an adjuvant in a concentration of about 0.1mg/ml to about 1.0mg/ml, 0.1mg/ml to about 0.9mg/ml, 0.1mg/ml to about 0.8mg/ml, 0.1mg/ml to about 0.7mg/ml, 0.1mg/ml to about 0.6mg/ml, 0.1mg/ml to about 0.5mg/ml, 0.1mg/ml to about 0.4mg/ml, 0.1mg/ml to about 0.3mg/ml, 0.1mg/ml to about 0.2mg/ml, 0.25mg/ml to about 0.95mg/ml, 0.25mg/ml to about 0.85mg/ml, 0.25mg/ml to about 0.75mg/ml, 0.25mg/ml to about 0.65mg/ml, 0.25mg/ml to about 0.55mg/ml, 0.45mg/ml to about 0.35mg/ml, 0.35mg/ml, 0.5mg/ml to about 1.0mg/ml, 0.5mg/ml to about 0.9mg/ml, 0.5mg/ml to about 0.8mg/ml, 0.5mg/ml to about 0.75mg/ml, 0.5mg/ml to about 0.7mg/ml, 0.5mg/ml to about 0.65mg/ml, 0.5mg/ml to about 0.6mg/ml, 0.75mg/ml to about 1.0mg/ml, 0.75mg/ml to about 0.95mg/ml, 0.75mg/ml to about 0.9mg/ml, and 0.75mg/ml to about 0.85 mg/ml. In a preferred embodiment, the immunogenic composition comprises the adjuvant at a concentration of about 0.25mg/ml to about 0.75mg/ml, most preferably about 0.5 mg/ml.
In a preferred embodiment, the adjuvant is an aluminum-based adjuvant at a concentration of about 0.5 mg/ml. In one such embodiment, the aluminum-based adjuvant is aluminum phosphate or aluminum hydroxyphosphate.
In one embodiment, the immunogenic composition comprises a polysaccharide-protein conjugate as described herein, a buffering agent, a surfactant, an excipient, and optionally an adjuvant, wherein the composition is buffered to a pH of about 6.0 to about 7.0.
In one such embodiment, the immunogenic composition comprises a GBS polysaccharide-protein conjugate, a buffer, a surfactant, an excipient, and optionally an adjuvant, wherein the composition is buffered to a pH of about 6.0 to about 7.0, and wherein the capsular polysaccharide has a sialic acid level of greater than about 60%.
In particular embodiments, the immunogenic composition comprises a GBS polysaccharide-protein conjugate, histidine, polysorbate-80, sodium chloride, and optionally aluminum phosphate, wherein the composition is buffered to a pH of about 6.0 to about 7.0, and wherein the capsular polysaccharide has a sialic acid level of greater than about 60%.
In a preferred embodiment, the immunogenic composition comprises about 5 to about 50mcg/ml of GBS polysaccharide-protein conjugate, about 10 to about 25mM histidine, about 0.01 to about 0.03% (v/w) polysorbate-80, about 10 to about 250mM sodium chloride, and optionally about 0.25 to about 0.75mg/ml aluminum as aluminum phosphate, wherein the capsular polysaccharide has a sialic acid level of greater than about 60%.
In one such embodiment, the immunogenic composition comprises at least two GBS polysaccharide-protein conjugates, a buffer, a surfactant, an excipient, and optionally an adjuvant, wherein the composition is buffered to a pH of about 6.0 to about 7.0, and wherein the conjugate comprises a capsular polysaccharide from Group B Streptococcus (GBS) serotype IV and at least one additional serotype selected from the group consisting of: ia. Ib, II, III, V, VI, VII, VIII and IX.
In particular embodiments, the immunogenic composition comprises at least two GBS polysaccharide-protein conjugates, histidine, polysorbate-80, sodium chloride, and optionally aluminum phosphate, wherein the composition is buffered to a pH of about 6.0 to about 7.0, and wherein the conjugate comprises a capsular polysaccharide from Group B Streptococcus (GBS) serotype IV and at least one additional serotype selected from the group consisting of: ia. Ib, II, III, V, VI, VII, VIII and IX.
In a preferred embodiment, the immunogenic composition comprises at least two GBS polysaccharide-protein conjugates of about 5mcg/ml to about 50mcg/ml each, about 10mM to about 25mM histidine, about 0.01% to about 0.03% (v/w) polysorbate-80, about 10mM to about 250mM sodium chloride, and optionally about 0.25mg/ml to about 0.75mg/ml aluminum as aluminum phosphate, wherein the conjugate comprises a capsular polysaccharide from Group B Streptococcus (GBS) serotype IV and at least one additional serotype selected from the group consisting of: ia. Ib, II, III, V, VI, VII, VIII and IX.
Evaluation of immunogenic compositions
Various in vitro assays are used to assess the immunogenicity of the immunogenic compositions of the invention. For example, a mixture of streptococcal cells, heat-inactivated serum containing specific antibodies to the antigen under study, and an exogenous complement source are incubated together for in vitro opsonin analysis. Opsonophagocytosis is performed during incubation of freshly isolated polymorphonuclear cells (PMNs) or differentiated effector cells such as HL60 and antibody/complement/streptococcal cell mixtures. Upon opsonophagocytosis, bacterial cells coated with antibodies and complement are killed. Colony forming units (cfu) of surviving bacteria recovered from opsonophagocytosis were determined by plating the assay mixture. The reported titer is the reciprocal of the highest dilution that resulted in 50% of the bacteria being killed, as determined by comparison to the assay control group.
Whole cell ELISA assays can also be used to assess the in vitro immunogenicity and surface exposure of antigens, where the target strain (streptococcus agalactiae) is plated on a plate (e.g., a 96-well plate) and test sera from immunized animals are reacted with bacterial cells. If an antibody specific for the test antigen reacts with a surface-exposed epitope of the antigen, this can be detected by standard methods known to those skilled in the art. Alternatively, flow cytometry can be used to measure surface exposure of capsular polysaccharide antigens and specificity of antibodies (including monoclonal antibodies).
Antigens that exhibit the desired in vitro activity can then be tested in an in vivo animal challenge model. In some embodiments, the animal (e.g., mouse) is immunized with the immunogenic composition by immunization methods and routes known to those of skill in the art (e.g., intranasal, parenteral, oral, rectal, vaginal, transdermal, intraperitoneal, intravenous, subcutaneous, etc.). After immunization of animals with GBS immunogenic compositions, the animals were challenged with streptococcus agalactiae strains and analyzed for resistance to streptococcal infection.
In one embodiment, a pathogen-free mouse is immunized and challenged with Streptococcus agalactiae. For example, mice are immunized with one or more doses of the antigen of interest in the immunogenic composition. Subsequently, mice were challenged with streptococcus agalactiae and survival was monitored over time after challenge.
Application method
As used herein, "immune function impaired" refers to a subject suffering from a defect in the cellular and/or humoral arm (cellular and/or humoral arm) of the immune system. Thus, the extent of immune function deficiency is considered to range from mild attenuation during immunization to complete immunosuppression.
The term "subject" refers to a mammal, bird, fish, reptile (reptile), or any other animal. The term "subject" also includes humans. The term "subject" also includes household pets. Non-limiting examples of household pets include: dogs, cats, pigs, rabbits, rats, mice, gerbils, hamsters, guinea pigs, ferrets, birds, snakes, lizards, fish, turtles, and frogs. The term "subject" also includes livestock. Non-limiting examples of livestock include: alpaca, bison, camel, cattle, deer, pig, horse, llama, mule, donkey, sheep, goat, rabbit, reindeer, yak, chicken, goose and turkey.
As used herein, "treatment" (including variations thereof, such as "treating" or "therapy") refers to any one or more of the following: (i) preventing infection or reinfection, as in a traditional vaccine, (ii) reducing the severity of symptoms or eliminating symptoms, and (iii) substantially or completely eliminating the pathogen or condition of interest. Thus, treatment can be effected prophylactically (pre-infection) or therapeutically (post-infection). In the present invention, either prophylactic or therapeutic treatment may be used. According to a particular embodiment of the present invention, compositions and methods are provided for treating (including prophylactically and/or therapeutically immunizing) a host animal against an antimicrobial infection (e.g., a bacterium, such as streptococcus agalactiae). The methods of the invention are useful for conferring prophylactic and/or therapeutic immunity to a subject. The methods of the invention can also be performed in a subject for biomedical research applications.
In another aspect, the invention relates to a method of inducing an immune response to GBS in a subject by administering to the subject an effective amount of an immunogenic composition described herein. In one embodiment, the invention relates to a method of preventing or alleviating a group B streptococcus-associated disease or condition in a subject by administering to the subject an effective amount of an immunogenic composition described herein. In one aspect, the invention relates to the use of an immunogenic composition as described herein as a medicament. In one aspect, the invention relates to the use of an immunogenic composition described herein in a method of eliciting an immune response to GBS in a subject. In particular embodiments, the subject is a female intended to be pregnant or a pregnant female. In one such embodiment, the pregnant female is third trimester second half of its pregnancy, such as at least at 20 weeks gestation or at least at 27 weeks gestation. In a preferred embodiment, the pregnant female is between 27 and 36 weeks gestation. In another embodiment, the subject is an elderly adult such as an adult of 50 years or older, an adult of 65 years or older, and an adult of 85 years or older. In another embodiment, the subject is immunocompromised. In one aspect, the subject has a medical condition selected from the group consisting of: obesity, diabetes, HIV infection, cancer, cardiovascular disease or liver disease. In a preferred embodiment, the group B streptococcus is streptococcus agalactiae (s.
In one embodiment, the immunogenic composition comprises a polysaccharide-protein conjugate comprising GBS serotype IV and at least one additional serotype selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In another embodiment, the conjugate comprises GBS serotype IV and at least two additional serotypes selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In further embodiments, the conjugate comprises GBS serotype IV and at least three further serotypes selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In another embodiment, the conjugate comprises GBS serotype IV and at least four additional serotypes selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In particular embodiments, the conjugates comprise GBS serotypes Ia, Ib, II, III, and IV. In another embodiment, the conjugate comprises GBS serotype IV and at least five additional serotypes selected from the group consisting of: serotypes Ia, Ib, II, III, V, VI, VII, VIII and IX. In another embodiment, the composition comprises GBS serotype V. In particular embodiments, the conjugates comprise GBS serotypes Ia, Ib, II, III, and V. In a preferred embodiment, the immunogenic composition comprises six polysaccharide-protein conjugates from GBS serotypes Ia, Ib, II, III, IV and V. One aspect of the invention relates to immunogenic compositions that do not have immune interference.
The immunogenicity or effective amount of the immunogenic composition can be determined by performing a dose response study in which subjects are immunized with escalating amounts of the immunogenic composition and the immune response is analyzed to determine the optimal dose. The starting point of the study can be inferred from the immunization data in the animal model. The dosage may vary depending on the particular circumstances of the subject. This amount can be determined in routine tests by means known to the person skilled in the art.
An immunologically effective amount of the immunogenic composition is administered to the subject in an appropriate number of doses to elicit an immune response. The dosage may vary depending on the particular condition of the subject, such as age and weight. This amount can be determined in routine tests by means known to the person skilled in the art.
In one embodiment, a patient administered an immunogenic composition of the invention exhibits a reduced streptococcus agalactiae bacterial load (carriage rate). Such a reduction in the rate of carrier or an extension of the time interval as a non-carrier after administration of the immunogenic composition is significant from a medical need point of view. For example, a reduction in the overall streptococcus agalactiae carrier's bacterial load rate can be assessed following administration of a dose of an immunogenic composition of the invention. For example, a panel of adults aged 18 to 50 years may be screened for carriage (carriage) by nasal, laryngeal, axillary, rectal, perineal and vaginal swabs (swab) 1 day prior to administration of the immunogenic composition, followed by culture to determine carriage status. Next, the group was administered with the immunogenic composition of the invention, while the other group received a control. Nasal, laryngeal, axillary, rectal, perineal and vaginal swabs were performed continuously weekly for 12 weeks after administration of the immunogenic composition and once a month for up to 6 months and compared to placebo. One primary endpoint is a comparison of the germ carrying rate (carriage rate) of patients following administration of the immunogenic composition and placebo, at a period of 3 months after immunization.
Antibodies
An "antibody" is an immunoglobulin molecule that is capable of specifically binding to a target (such as a carbohydrate, polynucleotide, lipid, polypeptide, etc.) through at least one antigen recognition site located in the variable region of the immunoglobulin molecule. Unless the context indicates otherwise, this term as used herein is intended to include not only intact polyclonal or monoclonal antibodies, but also genetically engineered antibodies (e.g., chimeric, humanized and/or derivatized to alter effector function, stability and other biological activity) and fragments thereof (such as Fab, Fab ', F (ab')2, Fv), single chain (ScFv) and domain antibodies, including shark and camelid antibodies), and fusion proteins comprising an antibody portion, multivalent antibodies, multispecific antibodies (e.g., bispecific antibodies, so long as they exhibit the desired biological activity), and antibody fragments as described herein, as well as any other modified configuration of the immunoglobulin molecule that includes an antigen recognition site. Antibodies include any class of antibody, such as IgG, IgA, or IgM (or subclasses thereof), and the antibody need not be of any particular class. Immunoglobulins can be assigned to different classes depending on the amino acid sequence of the constant domain of the antibody heavy chain. There are five major classes of human immunoglobulins: IgA, IgD, IgE, IgG and IgM, some of which may be further classified into subclasses (isotypes), such as IgG1, IgG2, IgG3, IgG4, IgA1 and IgA 2. The heavy chain constant domains corresponding to different classes of immunoglobulins are referred to as α, δ, ε, γ, and μ, respectively. The subunit structures and three-dimensional configurations of different classes of immunoglobulins are well known.
An "antibody fragment" includes only a portion of an intact antibody, wherein the portion preferably retains at least one, and preferably most or all, of the functions normally associated with the portion present in an intact antibody.
As used herein, the "functional activity" of an antibody or "functional antibody" means that the antibody specifically binds at least to an antigen. Other functions are known in the art and may include other components of the immune system that can scavenge or kill pathogens, such as through opsonization, ADCC, or complement-mediated cytotoxicity. After antigen binding, any subsequent antibody function may be mediated through the Fc region of the antibody. Antibody opsonophagocytosis assay (OPA) is an in vitro assay designed to measure in vitro Ig complement-assisted bacterial killing by effector cells (leukocytes), thereby mimicking biological processes. Antibody binding may also directly inhibit the biological function of the antigen to which it binds. In some embodiments, a "functional antibody" refers to an antibody that is functional as measured by killing bacteria in an animal efficacy model or an opsonophagocytic killing assay that demonstrates that the antibody kills bacteria.
In one aspect, the invention relates to an isolated antibody or fragment thereof that specifically binds to a polysaccharide described herein. An "isolated" antibody, as used herein, refers to an antibody that has been identified and separated and/or recovered from a component of its natural environment. Contaminating components of their natural environment are substances that would interfere with diagnostic or therapeutic uses of the antibody, and may include enzymes, hormones, and other proteinaceous or nonproteinaceous solutes. In exemplary embodiments, the antibody will be purified (1) to greater than 95% by weight of the antibody (as determined by the Lowry method), most preferably greater than 99% by weight, (2) to an extent sufficient to obtain at least 15 residues of the N-terminal or internal amino acid sequence by using a rotor-cup sequencer, or (3) to be homogeneous when analyzed by SDS-PAGE using coomassie blue or, preferably, silver staining under reducing or non-reducing conditions. Isolated antibodies include antibodies in situ within recombinant cells, as at least a component of the antibody's natural environment will not be present. However, an isolated antibody will typically be produced by at least one purification step.
An antibody that "specifically binds" or is "specific for" a particular polysaccharide or an epitope on a particular polysaccharide is an antibody that binds to the particular polysaccharide or epitope on a particular polysaccharide without substantially binding to any other polysaccharide or polysaccharide epitope.
As used herein, "label" refers to a detectable compound or composition that is conjugated, directly or indirectly, to an antibody, thereby producing a "labeled" antibody. The label may be detectable by itself (e.g., radioisotope labels or fluorescent labels) or, in the case of an enzymatic label, may catalyze chemical alteration of a substrate compound or composition that is detectable.
The invention further provides antibodies and antibody compositions that specifically and selectively bind to one or more antigens of the immunogenic compositions of the invention. In some embodiments, the antibodies are produced upon administration of the immunogenic compositions of the invention to a subject. In some embodiments, the invention provides purified or isolated antibodies against one or more antigens of the immunogenic compositions of the invention. In some embodiments, the antibodies of the invention are functional as measured by killing bacteria in an animal efficacy model or via an opsonophagocytic killing assay. In some embodiments, the antibodies of the invention confer passive immunity to a subject. The invention further provides polynucleotide molecules encoding the antibodies or antibody fragments of the invention, and cells or cell lines (such as hybridoma cells or other genetically engineered cell lines for recombinant production of antibodies) and transgenic animals (using techniques well known to those skilled in the art) that produce the antibodies or antibody compositions of the invention.
The antibodies or antibody compositions of the invention are useful for treating or preventing a streptococcus infection, disease or condition associated with streptococcus agalactiae in a subject, the method comprising producing a polyclonal or monoclonal antibody preparation, and using the antibodies or antibody compositions to confer passive immunity to the subject. The antibodies of the invention may also be used in diagnostic methods, such as detecting the presence or quantification of one or more antigens of the immunogenic compositions of the invention.
Antibody responses to repetitive structures (e.g., the polysaccharides of the invention) may exhibit some unique properties. For example, the regularity of the repeating units may mean that antigenic molecules of very different molecular weights can bind to antibodies specific for the polysaccharide. Second, the longer polysaccharide repeat structure is capable of inducing T cell-independent antibody responses. Thus, both T cell independent and T cell dependent antibody responses can be stimulated when using polysaccharides conjugated to protein carriers with T helper epitopes. Thus, the immune response can be modified by appropriate choice of polysaccharide size and whether or not a carrier protein is used.
Polyclonal antibodies
In certain embodiments, the anti-polysaccharide antibody is a polyclonal antibody. Polyclonal antibodies as defined herein refers to a mixture of antibodies derived from serum preparations and from different B cell clones with different specificities. Methods for the preparation and characterization of polyclonal antibodies are known in the art.
Polyclonal antibodies are raised in a subject (e.g., a mammal) by administration of one or more immunogens or immunogenic compositions described herein and, if desired, injections of adjuvants, buffers, and/or diluents. There are many animal species that can be used to make specific antisera. Generally, the animal used for the production of the anti-sugar polyclonal antiserum is a non-human primate, goat, sheep, rabbit, mouse, rat, hamster, or guinea pig. Typically, the immunogen or immunogenic composition with or without adjuvant is injected into the mammal by multiple injections. The immunogenic material may include a polysaccharide, an oligosaccharide, a polysaccharide as described herein, a polysaccharide-protein conjugate, or a larger immunogenic assembly. Typically, blood is collected from the immunized animal, allowed to clot and serum is harvested, starting 2 to 6 weeks after the first immunization. The serum contains anti-sugar polyclonal antibodies from the immunized animal, commonly referred to as antisera.
Monoclonal antibodies
Anti-sugar monoclonal antibodies can be made by using known hybridoma techniques. Generally, the manufacture of monoclonal antibodies involves first immunizing a suitable target animal host with a selected immunogen comprising a polysaccharide, oligosaccharide, polysaccharide or polysaccharide-protein conjugate of the invention. Adjuvants, buffers and/or diluents may be included if desired. The immunization is carried out in a manner sufficient to induce B lymphocytes to make or express antibodies that specifically bind to the polysaccharide or conjugate thereof. Alternatively, lymphocytes are immunized in vitro.
The lymphocytes are then fused with an immortalized cell line using a suitable fusing agent, such as polyethylene glycol, to form hybridoma cells. The source of the lymphocytes determines that the monoclonal antibody is of human or animal origin. Generally, peripheral blood lymphocytes ("PBLs") are used if antibodies and cells of human origin are desired, and spleen cells or lymph node cells are used if non-human mammalian sources are desired.
Immortalized cell lines are generally transformed mammalian cells, in particular myeloma cells of rodent, bovine and human origin. Typically, rat or mouse myeloma cell lines are used. The hybridoma cells are cultured in a suitable medium, preferably containing one or more substances that inhibit the growth or survival of the unfused, immortalized cells. For example, if the parental cells lack the enzyme hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT), the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine ("HAT medium"), which substances will prevent the growth of HGPRT-deficient cells.
Immortalized cell lines are selected based on practical considerations such as the species of origin, fusion and growth characteristics. For example, suitable immortalized cell lines are those that fuse efficiently, support stable and high level expression of antibodies by the selected antibody-producing cells, and are sensitive to a culture medium (such as HAT medium). Examples of immortalized cell lines include: murine myeloma lines. Human myeloma and mouse-human heteromyeloma cell lines have also been described for the production of human monoclonal antibodies.
Monoclonal antibodies are secreted into the culture medium by the hybridoma cells. The medium is then analyzed for the presence of monoclonal antibodies that recognize and bind to the polysaccharide. The anti-polysaccharide binding specificity of a particular monoclonal antibody produced by a hybridoma cell is determined by one of a number of procedures well known in the art. For example, antibody binding specificity can be determined by immunoprecipitation, Radioimmunoassay (RIA), Western blotting, enzyme-linked immunosorbent assay (ELISA), or surface plasmon resonance (e.g., Biacore). The precise epitope recognized by a monoclonal antibody is determined by epitope mapping. Such techniques and assays are well known in the art.
After identification of antibody-producing hybridoma cells with the desired specificity, the clonal strain is subcloned by limiting dilution and cultured using standard methods. Suitable media for this purpose include, for example, Dulbecco's Modified Eagle's Medium and RPMI-1640 Medium. Alternatively, the hybridoma cells are grown in vivo in a mammal as ascites fluid. Monoclonal antibodies secreted by the subclones are isolated or purified from the culture medium or ascites fluid by conventional immunoglobulin purification procedures such as, for example, protein a-sepharose, hydroxyapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
Alternatively, antibodies having the desired specificity and derived from the desired source species can be obtained by using phage display libraries. Furthermore, examples of methods and reagents particularly suitable for generating and screening antibody display libraries can be found in the art.
Antibody uses
In one aspect, the invention relates to the use of an immunogenic composition as described herein for the manufacture of GBS antibodies and/or antibody fragments. The polysaccharide-protein conjugates described herein and/or antibodies produced therefrom can be used in a variety of immunodiagnostic techniques known to those skilled in the art, including ELISA and microarray-related techniques. In addition, these reagents can be used to assess antibody responses, including, for example, serum antibody levels against immunogenic polysaccharide conjugates. The assay methodology of the present invention may involve the use of a label such as a fluorescent, chemiluminescent, radioactive, enzymatic label, or dye molecule, and/or a second immunoreagent for the direct or indirect detection of the complex between an antigen or antibody in the biological sample and a corresponding antibody or antigen bound to a solid support.
The antibodies or antibody fragments produced may also be used in passive immunotherapy or prophylactic therapy against streptococcal infection.
Method for producing polysaccharides
In another aspect, the invention relates to a method for making the polysaccharides described herein. The method comprises culturing GBS and collecting the polysaccharide produced by the bacterium. In one embodiment, the GBS comprises streptococcus agalactiae. The bacterium may be any strain of streptococcus agalactiae. In a preferred embodiment, the bacterium is a coated strain of Streptococcus agalactiae (encapsulated strain). Streptococcus agalactiae strains for use in the present invention include 090, A909(ATCC accession number BAA-1138), 515(ATCC accession number BAA-1177), B523, CJB524, MB4052(ATCC accession number 31574), H36B (ATCC accession number 12401), S40, S42, MB 4053(ATCC accession number 31575), M709, 133, 7357, PFEGBST0267, MB 4055(ATCC accession number 31576), 18RS21(ATCC accession number BAA-1175), S16, S20, V8(ATCC accession number 12973), DK21, DK23, UAB, 5401, PFEGBST0708, MB 4082(ATCC accession number 31577), M132, UA 110, M781(ATCC accession number BAA-22), D136D 2 (3) (ATCC accession number NI 03), M782, S23, 120, MB 4316 (ATCC accession number BAA-732), ATCC accession number 314933, ATCC accession number 319, ATCC accession number CJA-685 (ATCC accession number CJA-1179), ATCC accession number CJ-685-6853, ATCC accession number CJ-685 (ATCC accession number 319, ATCC accession number 3139, ATCC accession number CJ-685-6853), ATCC accession number 97, ATCC accession number CJ-9, ATCC accession number 319, ATCC accession number CJ-6853 (ATCC accession number 97) and ATCC accession number 319, ATCC accession number 31577), and ATCC accession number 319, ATCC accession number 31577, ATCC accession number 319, CJB112, 2603V/R (ATCC accession number BAA-611), NCTC 10/81, CJ11, PFEGBST0837, 118754, 114852, 114862, 114866, 118775, B4589, B4645, SS1214, CZ-PW-119, 7271, CZ-PW-045, JM 3069130013, JM 9172, IT-NI-016, IT-PW-62 and IT-PW-64.
The polysaccharides described herein can be produced by culturing GBS in an appropriate medium. Suitable media may include Columbia broth. The culture medium may comprise dextrose, hemin, and/or glucose. Preferably, the medium comprises Columbia broth and dextrose. If Streptococcus agalactiae is cultured using Columbia broth and dextrose, the culture temperature is preferably 20 to 40 ℃, preferably 37 ℃. In a preferred embodiment, the bacteria are cultured under aerobic conditions. In another preferred embodiment, the bacteria are cultured for 12 to 60 hours.
The polysaccharide can be collected from the resulting culture using methods known in the art for collecting the target substance from the culture, such as, for example, heating, enzymatic treatment, centrifugation, precipitation, treatment with activated carbon, and/or filtration. (see, e.g., U.S. patent application publication Nos. 2006/0228380, 2006/0228381, 2007/0184071, 2007/0184072, 2007/0231340, and 2008/0102498; International patent application publication No. WO 2008/118752). In one embodiment, the culture containing the bacteria and the polysaccharide is centrifuged and treated with an enzyme, such as, for example, lysozyme, ribonuclease, deoxyribonuclease, pronase, mutanolysin, and combinations thereof. For example, in one embodiment, a suitable organic solvent is added to the obtained supernatant to precipitate the protein, and the precipitate is removed by centrifugation. Then, the polysaccharide is precipitated by further adding an appropriate organic solvent to the supernatant and the polysaccharide can be collected by centrifugation. More specifically, the polysaccharides described herein can be obtained by the following steps: ethanol was added to the supernatant from which the bacteria had been removed to a final concentration of about 25% by volume, the precipitate containing the protein was removed by centrifugation, ethanol was further added thereto to a final concentration of about 75% by volume, and then the precipitate was collected by centrifugation. The resulting precipitate can be dried with nitrogen. The resulting pellet was resuspended in Tris and 0.05% sodium azide.
Another aspect of the invention provides novel methods of using organic reagents, such as derivatized hydroxylamine compounds, to isolate the majority of intact high molecular weight CP, while retaining the N-and O-acetyl groups. Since this method does not lyse the cells, CP separated by centrifugation is minimally contaminated with intracellular components and can result in higher overall yields. In addition, since the group B antigen impurities have multiple phosphodiester bonds, these agents cleave the group B antigen impurities into very small fragments that can be easily removed by diafiltration.
In one embodiment, the CP is isolated by reacting hydroxylamine with a cell paste comprising capsular polysaccharide producing bacteria. In certain embodiments, the method further comprises a centrifugation step. In another embodiment, the method further comprises a filtration step. In another embodiment, the capsular polysaccharide producing bacterium is selected from the group consisting of: streptococcus agalactiae, Streptococcus pneumoniae, Staphylococcus aureus, Neisseria meningitidis, Escherichia coli, Salmonella typhi, Haemophilus influenzae, Klebsiella pneumoniae, enterococcus faecium, and enterococcus faecalis.
In one aspect of the invention, the hydroxylamines may be those listed in table 2 in example 2. In a preferred embodiment, the hydroxylamine is selected from the group consisting of: dibenzylhydroxylamine; diethylhydroxylamine; a hydroxylamine; ethylene diamine; triethylenetetramine; 1,1,4,7,10,10 hexamethyl triethylenetetramine; and 2,6,10, trimethyl 2,6,10 triazaundecane.
In one aspect of the invention, the concentration of hydroxylamine is about 5mM to about 200mM, such as about 5mM to about 150mM, about 5mM to about 100mM, about 5mM to about 75mM, about 5mM to about 50mM, about 5mM to about 25mM, about 5mM to about 10mM, 10mM to about 200mM, such as about 10mM to about 150mM, about 10mM to about 100mM, about 10mM to about 75mM, about 10mM to about 50mM, about 10mM to about 25mM, about 25mM to about 200mM, about 25mM to about 150mM, about 25mM to about 100mM, about 25mM to about 75mM, about 25mM to about 50mM, about 50mM to about 200mM, about 50mM to about 150mM, 50mM to about 100mM, and about 50mM to about 75 mM.
In another aspect, the pH of the reaction is maintained at about 5.5 to about 9.5, such as about 5.5 to about 9.0, about 5.5 to about 8.5, about 5.5 to about 8.0, about 5.5 to about 7.5, about 5.5 to about 7.0, about 5.5 to about 6.5, about 6.0 to about 9.5, about 6.0 to about 9.0, about 6.0 to about 8.5, about 6.0 to about 8.0, about 6.0 to about 7.5, about 6.0 to about 7.0, about 6.5 to about 9.5, about 6.5 to about 8.5, about 6.5 to about 8.0, about 6.5 to about 7.5, about 7.0 to about 9.5, about 7.0 to about 9.0, 7.0 to about 8.5, and about 7.0 to about 8.0.
In another aspect of the invention, the reaction occurs at a temperature of about 20 ℃ to about 85 ℃, such as about 20 ℃ to about 80 ℃, about 20 ℃ to about 75 ℃, about 20 ℃ to about 70 ℃, about 20 ℃ to about 65 ℃, about 20 ℃ to about 60 ℃, about 20 ℃ to about 55 ℃, about 20 ℃ to about 50 ℃, about 25 ℃ to about 85 ℃, about 25 ℃ to about 80 ℃, about 25 ℃ to about 75 ℃, about 25 ℃ to about 70 ℃, about 25 ℃ to about 65 ℃, about 25 ℃ to about 60 ℃, about 25 ℃ to about 55 ℃, about 25 ℃ to about 50 ℃, about 30 ℃ to about 85 ℃, about 30 ℃ to about 80 ℃, about 30 ℃ to about 75 ℃, about 30 ℃ to about 70 ℃, about 30 ℃ to about 65 ℃, about 30 ℃ to about 60 ℃, about 30 ℃ to about 55 ℃, about 30 ℃ to about 50 ℃, about 35 ℃ to about 85 ℃, about 35 ℃ to about 80 ℃, about 35 ℃ to about 75 ℃, about 35 ℃ to about 70 ℃, about 35 ℃ to about 65 ℃, about 35 ℃ to about 60 ℃, about 35 ℃ to about 60 ℃. (C.) About 35 ℃ to about 55 ℃, about 40 ℃ to about 85 ℃, about 40 ℃ to about 80 ℃, about 40 ℃ to about 75 ℃, about 40 ℃ to about 70 ℃, about 40 ℃ to about 65 ℃, about 40 ℃ to about 60 ℃, about 45 ℃ to about 85 ℃, about 45 ℃ to about 80 ℃, about 45 ℃ to about 75 ℃, about 45 ℃ to about 70 ℃, about 45 ℃ to about 65 ℃, about 50 ℃ to about 85 ℃, about 50 ℃ to about 80 ℃, about 50 ℃ to about 75 ℃, about 50 ℃ to about 70 ℃, about 55 ℃ to about 85 ℃, about 55 ℃ to about 80 ℃, about 55 ℃ to about 75 ℃, about 60 ℃ to about 85 ℃ and about 65 ℃ to about 85 ℃.
In another aspect, the reaction time is from about 10 hours to about 90 hours, such as from about 10 hours to about 85 hours, from about 10 hours to about 80 hours, from about 10 hours to about 75 hours, from about 10 hours to about 70 hours, from about 10 hours to about 60 hours, from about 10 hours to about 50 hours, from about 10 hours to about 40 hours, from about 10 hours to about 30 hours, from about 10 hours to about 25 hours, from about 10 hours to about 20 hours, from about 10 hours to about 15 hours, from about 15 hours to about 90 hours, from about 15 hours to about 85 hours, from about 15 hours to about 80 hours, from about 15 hours to about 75 hours, from about 15 hours to about 70 hours, from about 15 hours to about 60 hours, from about 15 hours to about 50 hours, from about 15 hours to about 40 hours, from about 15 hours to about 30 hours, from 15 hours to about 20 hours, such as from about 20 hours to about 90 hours, from about 20 hours to about 85 hours, from about 20 hours to about 80 hours, About 20 hours to about 75 hours, about 20 hours to about 70 hours, about 20 hours to about 60 hours, about 20 hours to about 50 hours, about 20 hours to about 40 hours, about 20 hours to about 30 hours, and about 20 hours to about 25 hours.
Alternatively, in another embodiment of the invention, the polysaccharide is chemically synthesized. The polysaccharide can be synthesized chemically according to a conventional method.
In another embodiment of the invention, the polysaccharide is produced by expression after cloning and expressing the biosynthetic pathway for producing the polysaccharide in an alternative host. For example, a host cell can be modified to produce a polysaccharide having a structure similar to the polysaccharide described herein, wherein the repeat units of the polysaccharide produced in the host cell are in partial agreement with the repeat units of the polysaccharide described herein. For example, a polysaccharide is structurally similar to a polysaccharide described herein if its repeat units have missing branches, are heterogeneous in size, and/or are heterogeneous in a branched structure (a branching arrangement), as compared to the repeat units of the polysaccharide described herein. Preferably, the host cell is a bacterial host cell.
Examples
The following examples illustrate some embodiments of the invention. It is to be understood, however, that these examples are for illustrative purposes only and are not intended to fully limit the conditions and scope of the present invention. It will be appreciated that when typical reaction conditions (e.g., temperature, reaction time, etc.) have been given, conditions above and below the specified ranges may also be used, although generally less convenient. All parts and percentages mentioned herein are by weight and all temperatures are expressed in degrees celsius unless otherwise indicated.
Furthermore, unless otherwise specified, the following examples are carried out using standard techniques well known and conventional to those skilled in the art. As noted above, the following examples are intended to be illustrative and should not be construed as limiting the scope of the invention in any way.
Example 1: manufacture of polysaccharide-protein conjugates with de-O-acetylated polysaccharides
The Streptococcus agalactiae strains of the individual serotypes are fermented in submerged culture (submerged culture) in a defined medium with controlled pH. The procedure and the culture medium used were optimized experimentally and were an extension of the basic technique described previously in von Hunolstein, C. et al, appl. Micro. Biotech.38(4):458-. The capsular polysaccharide is removed from the cells by NaOH treatment. After clarification (clarification), a series of UF/DF, precipitation and carbon filtration (carbon filtration) steps were performed to yield the purified polysaccharide. See, for example, U.S. patent No. 8,652,480. Use of reductive amination chemistry to react activated polysaccharide with CRM197And (6) conjugation. See, for example, U.S. patent No. 5,360,897.
Example 2 isolation of O-acetylated polysaccharides
The cell paste from GBS Capsular Polysaccharide (CP) serotype Ia obtained after fermentation broth (1.2L) heat killing (heat kill) and centrifugation was resuspended in 175mL of 25mM potassium phosphate buffer (25mM, pH 6.9). The suspension was mixed with an aqueous solution of hydroxylamine O-sulfonic acid (aqueous hydroxylamine O-sulfonic acid solution) to a final concentration of 10 mM. The pH of the suspension was determined to be about 5.8. The suspension was stirred at 55 ℃ for 72 hours. Thereafter, the suspension was centrifuged at about 10,000rpm and the supernatant was collected. Supernatants containing crude cleaved cp were analyzed for molecular weight and yield. The remaining fraction was purified by diafiltration (diafiltration) using water for injection (WFI) using a 30kDa MWCO membrane. The molecular weight of the purified polysaccharide was further analyzed by size exclusion chromatography (size exclusion chromatography) in combination with a multi-angle light scattering detector (SEC-MALS) (table 1).
TABLE 1 purification of GBS serotype Ia by diafiltration
Figure BDA0003350538080000881
Several hydroxylamines (both nitrogen and oxygen substituted compounds) were screened for activity using the method described above. The yield was calculated by gel permeation chromatography (gel permeation chromatography) of the raw supernatant (raw supernatant) in combination with multi-angle light scattering detection (GPC-MALS) using the Refractive Index (RI) reaction and the square of the specific refractive index increment (dn/dc) value of 0.135. The yield depends on the type of hydroxylamine and optimization of conditions such as concentration, temperature and reaction time (see table 2). Generally, increased hydroxylamine concentrations, higher temperatures and longer reaction times result in higher yields.
TABLE 2 screening of various hydroxylamines and Condition optimization of GBS capsular polysaccharide serotype Ia
Figure BDA0003350538080000891
Figure BDA0003350538080000901
Figure BDA0003350538080000911
Figure BDA0003350538080000921
Substituted and unsubstituted hydroxylamines were found to be very effective in releasing the GBS capsular polysaccharide from the cell wall. This method allows the isolation of high molecular weight CPS and the preservation of N-and O-acetyl groups. Of the several compounds screened, dibenzylhydroxylamine was considered to be the most effective. The data are shown in Table 3 ([ dibenzylhydroxylamine ] -50 mM; pH-7 to 8; temperature-50 ℃ C.; time-24 hours).
TABLE 3 GBS CP Release data Using dibenzylhydroxylamine
GBS type Ia Ib III
Polysaccharide release yield 86% 81% 46%
Total purification yield 63% 54% 30%
Molecular weight (Mw) 330kDa 212kDa 171kDa
O-acetylation (NMR) NA 31% 37%
N-acetyl (NMR) 106% 104% 87%
NA-serotype Ia is not O-acetylated
Due to the poor solubility of dibenzylhydroxylamine in water, alternative derivatives of hydroxylamine that are readily soluble in water and have similar or higher activity than dibenzylhydroxylamine are desirable. After screening some compounds, diethylhydroxylamine was considered a good choice. The data are shown in Table 4 ([ diethylhydroxylamine ] -100 mM; pH-7 to 8; temperature-60 ℃ C.; time-19 hours).
TABLE 4 GBS CP Release data Using Diethylhydroxylamine
Figure BDA0003350538080000922
Figure BDA0003350538080000931
Hydroxylamine (NH)2OH) has also been found to be effective in cleaving CPS from the cell wall. Data are shown in table 5 ([ hydroxylamine ]]-100 mM; pH-7 to 7.5; the temperature is-65 ℃; time-17 hours). In terms of serotype III, the yield after 17 hours was 54%; however, the yield increased to 70% after 3 days and half.
TABLE 5 GBS CP Release data Using hydroxylamine
Figure BDA0003350538080000932
Screening for oligoamines for release of GBS capsular polysaccharides from cells
Hydroxylamine and substituted compounds thereof have been found to be very effective in cleaving the capsular polysaccharide from the GBS cell wall. However, they were found to be less effective against serotypes II and V. Therefore, oligoamines (oligoamines) were tested because it is believed that they may be more active due to multiple amine functionalities.
Ethylene diamine (ethylenediamine) was found to be effective in releasing capsular polysaccharides from all serotypes. The data are shown in Table 6 ([ ethylenediamine ] -50 or 100 mM; pH 8.0; 16 h; 80 ℃; 25mM EDTA).
TABLE 6 GBS CP Release data Using ethylenediamine
GBS serotype Recovery of (%) Mw(kDa)
Ia 96% 242
Ib 83% 225
II 30% 76
III 68% 94
V 30% 235
Other representative oligoamines were tested for activity using serotype Ia and V cell pastes, but were also found to be less effective against serotype V. The data are shown in Table 7([ triethylenetetramine) ] -100 mM; pH-8.9; temperature-60 ℃ for-15 hours), Table 8([1,1,4,7,10,10 hexamethyltriethylenetetramine ] -10 mM; pH-6.3; temperature-60 ℃ for-20 hours) and Table 9([2,6,10, trimethyl 2,6,10 triazaundecane ] -10 mM; pH-7 to 8; temperature-60 ℃ for-19 hours).
TABLE 7 GBS CP Release data Using triethylenetetramine
Figure BDA0003350538080000941
GBS type Ia V
Polysaccharide Release yield% 100 2.5 days later-10
Molecular weight (Mw) 1280 nd
nd-not determined
TABLE 8 GBS CP Release data Using 1,1,4,7,10,10 hexamethyltriethylenetetramine
Figure BDA0003350538080000942
GBS type Ia V
Polysaccharide Release yield% 100 ~1%
Molecular weight (Mw) 980 nd
nd-not determined
TABLE 9 GBS CP Release data Using 2,6,10, trimethyl 2,6,10 triazaundecane
Figure BDA0003350538080000951
GBS type Ia V
Polysaccharide Release yield% 100 ~1%
Molecular weight (Mw) 1100 nd
nd-not determined
Example 3 conjugation of GBS capsular polysaccharides by reductive amination
Activated Polysaccharide (Activating Polysaccharide)
The polysaccharide oxidation reaction was carried out in 100mM potassium phosphate buffer (pH 6.0. + -. 0.5) by sequentially adding calculated amounts of 500mM potassium phosphate buffer (pH 6.0) and water for injection (WFI) to yield a final concentration of 2.0g/L of polysaccharide. The reaction pH was adjusted to about pH 6.0 if necessary. After adjusting the pH, the reaction temperature was adjusted to 23 ℃. The oxidation reaction was initiated by adding about 0.25 molar equivalents of sodium periodate. The oxidation reaction was carried out at 5. + -. 3 ℃ for about 16 hours.
The activated polysaccharide was concentrated and diafiltered (diafiltrations) using a 5K MWCO ultrafiltration cassette (5K MWCO ultrafiltration cassette). Diafiltration was performed against 20 diafiltration volumes (diavolme) of water for injection (WFI). The purified activated polysaccharide was then stored at 5 ± 3 ℃. Characterization of the purified activated polysaccharide can be determined by (i) colorimetric assay of sugar concentration, among others; (ii) determining the aldehyde concentration by colorimetric test; (iii) degree of oxidation (degree of oxidation); and (iv) determination of molecular weight by SEC-MALLS.
The degree of oxidation (DO ═ moles of saccharide repeat units/moles of aldehyde) of the activated polysaccharide was determined as follows:
The number of moles of saccharide repeat units is determined by various colorimetric methods, for example, by using the Anthrone (Anthrone) method, in which polysaccharides are first decomposed into monosaccharides by the action of sulfuric acid and heat. The anthrone reagent reacts with hexose (hexose) to form a yellow-green mixture whose absorbance (absorbance) is read spectrophotometrically at 625 nm. Within the analytical range, the absorbance is directly proportional to the amount of hexose present.
The moles of aldehyde were also determined simultaneously using MBTH colorimetry. The MBTH assay involves the formation of an azine (azine) compound by reacting the aldehyde group (from a given sample) with 3-methyl-2-benzothiazolone hydrazone (MBTH assay reagent). Excess 3-methyl-2-benzothiazolone hydrazone oxidizes to form a reactive cation (reactive cation). The reactive cation reacts with the azine to form a blue chromophore (chromophore). The formed chromophore was then read spectrophotometrically at 650 nm.
Mixing the activated polysaccharide with sucrose excipient and freeze drying
Will be activated at a rate of 25 grams of sucrose per gram of activated polysaccharideThe activated polysaccharide is mixed with sucrose. The bottle of the mixed mixture was then freeze dried. After freeze-drying, the bottles containing the freeze-dried activated polysaccharide were stored at-20 ± 5 ℃. CRM to be calculated 197The protein was shell-frozen (shell-freezen) and freeze-dried, respectively. Freeze-dried CRM197Store at-20. + -. 5 ℃.
Reconstitution of freeze-dried activated polysaccharide and carrier protein
The freeze-dried activated polysaccharide was reconstituted in anhydrous Dimethylsulfoxide (DMSO). When the polysaccharide was completely dissolved, an equal amount of anhydrous DMSO was added to the lyophilized CRM197To perform reconstruction (reconstruction).
Conjugation (Conjugating) and Capping (Capping)
Reconstituting the activated polysaccharide with reconstituted CRM197In a reaction vessel, they were combined and then mixed thoroughly to obtain a clear solution before starting conjugation with sodium cyanoborohydride (sodium cyanoborohydride). The final polysaccharide concentration in the reaction solution was about 1 g/L. The conjugation reaction was initiated by adding 1.0 to 1.5MEq of sodium cyanoborohydride to the reaction mixture and incubating at 23 ± 2 ℃ for 20 to 48 hours. By adding 2MEq of sodium borohydride (NaBH)4) Unreacted aldehyde is blocked to terminate the conjugation reaction. The capping reaction was allowed to proceed at 23. + -. 2 ℃ for 3. + -. 1 hours.
Purification of conjugates
In preparation for purification by tangential flow filtration (volumetric flow filtration) using a 100-K MWCO membrane, the conjugate solution was diluted 1:10 with cooled 5mM succinate-0.9% saline (pH 6.0).
The diluted conjugate solution was passed through a 5 μm filter and diafiltered using 5mM succinate/0.9% saline (pH 6.0) as the medium. After diafiltration was complete, the conjugate retentate (conjugate retentate) was transferred through a 0.22 μm filter. The conjugate was further diluted with 5mM succinate/0.9% saline (pH 6) to a target sugar concentration of about 0.5 mg/mL. Alternatively, the conjugate was purified by tangential flow filtration using 100 to 300KMWCO membranes using 20mM histidine-0.9% saline (pH 6.5). The final 0.22 μm filtration step was completed to obtain the immunogenic conjugate.
Example 4: altered conjugation conditions to GBS polysaccharide-CRM197Effect of the conjugate
GBS serotypes Ia, Ib, II, III, IV and V conjugates were generated by deliberate alteration of periodate oxidation/reductive amination chemistry (PO/RAC) conditions including solvent for reagents (aqueous medium versus DMSO), alteration of sialic acid levels and degree of oxidation/saccharide epitope modification in the starting polysaccharide. In general, conjugates made using DMSO as a solvent were found to have lower levels of unreacted (free) polysaccharide, higher conjugate molecular weight, and higher saccharide/protein ratio than conjugates made using aqueous media.
Conjugation methods that produce conjugates with lower levels of unreacted (free) polysaccharide are advantageous and preferred. It is well known that high levels of unreacted (free) polysaccharide may cause excessive T-cell independent immune responses (T-cell independent immune responses), which may dilute the T-cell dependent response (T-cell dependent response) produced by polysaccharide-protein conjugates, thereby reducing the immunogenic response due to the conjugates.
Selected GBS polysaccharides were chemically desialylated (see Chaffin, D.O, et al, J Bacteriol 187(13):4615-4626(2005)) by methods known in the art to generate conjugate variants (conjugate variants) to determine the effect of% desialylation on immunogenicity. Desialylation above about 40% (i.e., sialic acid levels less than about 60%) has a negative impact on immunogenicity.
Likewise, in most cases, a degree of oxidation of less than about 5, or more than about 20% of carbohydrate epitope modification, negatively affects immunogenicity. Since the oxidation reaction occurs via sialic acid on the capsular polysaccharide, the results appear to indicate that modification of the saccharide epitope by more than about 20% reduces the sialic acid content, which results in reduced immunogenicity.
In contrast, conjugates with various sugar/protein ratios or polysaccharide molecular weights can produce immunogenic responses in mice, suggesting that the range of accepted standards (acceptance criteria) for these properties is quite broad.
Alternative chemical routes are also used to generate additional conjugate variants (conjugate variants). An alternative chemistry involves reacting a polysaccharide with carbonyl bis-triazole (CDT) and performing a conjugation reaction in DMSO to generate a conjugate. In another alternative chemistry, the polysaccharide was oxidized by using TEMPO [ (2,2,6,6-Tetramethylpiperidin-1-yl) oxy ((2,2,6, 6-tetramethylpiperdin-1-yl) oxy) ] reagent (instead of sodium periodate) and then conjugated in DMSO using reductive amination chemistry (TEMPO/RAC) to generate the conjugate, as detailed in example 3 above. All conjugates generated by these alternative chemistries were demonstrated to be immunogenic in mice, suggesting the suitability of alternative chemical pathways other than PO/RAC. However, some conjugation chemistries perform better with certain serotypes than others.
OPA was performed according to Nanra, J.S., et al, hum.vaccine.Immunotherer, 9(3): 480-. After three doses (PD3) OPA titers (OPA titer) were provided as geometric means (geoman) from a group of 10 to 20 mice immunized at 1mcg/ml per dose of the respective conjugate.
GBS serotype Ia polysaccharide-CRM197Conjugates
Conjugates generated using PO/RAC and activated polysaccharide with DO of 16 to 17 (about 6% sugar epitope modification) showed immunogenicity (conjugates 1 and 3). However, the use of an activated polysaccharide with a DO of 5.4 (about 19% sugar epitope modification) had a negative impact on immunogenicity (conjugate 2). Likewise, 50% of the sialic acid levels produced little immunogenic response (conjugate 4). The results are shown in table 10.
TABLE 10 Process conditions for varying periodate Oxidation/reductive amination chemistry to GBS serotypes Ia-CRM197Effect of the conjugate
Figure BDA0003350538080000981
Figure BDA0003350538080000991
Additional conjugate variants were generated using alternative conjugation chemistries and conjugate molecular properties (results are shown in table 11). Conjugate 5 was generated by reacting the polysaccharide with carbonyl bis-triazole (CDT), and the conjugation reaction was performed in DMSO. Conjugate 6 was generated by oxidation of the polysaccharide using TEMPO reagent (instead of sodium periodate) followed by conjugation in DMSO using reductive amination chemistry, as detailed in example 3 above. Conjugate 7 was generated by PO/RAC and deliberately varying the conjugation parameters to produce conjugates with high sugar/protein ratios (SPR). Conjugate 8 was generated by PO/RAC using a polysaccharide with low MW (40 kDa). All these conjugates proved immunogenic in mice, which means the suitability of alternative (conjugation) conjugation chemistry in addition to periodate oxidation/reductive amination chemistry and alternative (conjugation) conjugate properties such as SPR and low MW of the original polysaccharide.
TABLE 11 Process conditions and alternative chemical selection for GBS serotypes Ia-CRM varying periodate Oxidation/reductive amination chemistry197Effect of the conjugate
Figure BDA0003350538080000992
Figure BDA0003350538080001001
GBS serotype Ib polysaccharide-CRM197Conjugates
In DMSO, conjugates generated using PO/RAC and activated polysaccharide with DO of 15.8 (about 6% sugar epitope modification) were shown to be immunogenic in mice (conjugates 9 and 11). When all other conjugate molecules were similar in character (conjugates 9 and 11, respectively), the conjugates generated by PO/RAC in DMSO were slightly more immunogenic than the conjugates generated by PO/RAC in aqueous media. However, the use of activated polysaccharide with a DO of 4.7 (about 21% sugar epitope modification) had a negative impact on immunogenicity (conjugate 10). In the conjugate (conjugate 12) generated using PO/RAC and 95% desialylated (5% sialic acid level) polysaccharide, immunogenicity was almost completely abolished with only minimal reaction. The results are shown in table 12.
TABLE 12 Process conditions for varying periodate Oxidation/reductive amination chemistry to GBS serotype Ib-CRM197Effect of the conjugate
Figure BDA0003350538080001002
Figure BDA0003350538080001011
Additional conjugate variants were generated using alternative conjugation chemistries and conjugation molecule properties (results are shown in table 13). Conjugate 13 was generated by PO/RAC using a polysaccharide with low sialylation (65%) in the starting polysaccharide (initial polysaccharide). Conjugate 14 was generated by PO/RAC and deliberately varying the conjugation parameters to produce a conjugate with a high sugar/protein ratio (SPR). Conjugate 15 was generated by reacting the polysaccharide with carbonyl bis-triazole (CDT) and the conjugation reaction was performed in DMSO. Conjugate 16 was generated by oxidation of the polysaccharide using TEMPO reagent (instead of sodium periodate) followed by conjugation in DMSO using reductive amination chemistry (TEMPO/RAC), as detailed in example 3 above. All these conjugates proved to be immunogenic in mice, which represents an alternative conjugation chemistry in addition to periodate oxidation/reductive amination chemistry, as well as the suitability of alternative conjugate properties such as SPR and low MW of the starting polysaccharide.
TABLE 13 Process for varying periodate Oxidation/reductive amination chemistryCondition and surrogate chemical selection for GBS serotype Ib-CRM197Effect of the conjugate
Figure BDA0003350538080001012
GBS serotype II polysaccharide-CRM197Conjugates
Conjugates generated using PO/RAC and activated polysaccharide with a DO of 4 to 15 (about 7 to 23% of saccharide epitope modification) were shown to be immunogenic in mice (conjugates 17 to 20). Conjugates generated using PO/RAC and a polysaccharide sialylated to 74% (26% desialylation) also showed immunogenicity (conjugate 20). The results are shown in Table 14.
TABLE 14 Process conditions for varying periodate Oxidation/reductive amination chemistry to GBS serotype II-CRM197Effect of the conjugate
Figure BDA0003350538080001021
Additional conjugate variants were generated using alternative conjugation chemistries and conjugation molecule properties (results are shown in table 15). Conjugates 21 and 22 were generated by PO/RAC and deliberately varying the conjugation parameters to produce conjugates with low and high saccharide/protein ratios (SPR), respectively. Conjugate 23 was generated by oxidation of the polysaccharide using TEMPO reagent (instead of sodium periodate) followed by conjugation in DMSO using reductive amination chemistry, as detailed in example 3 above. Conjugate 24 is generated by reacting the polysaccharide with carbonyl bis-triazole (CDT) and performing the conjugation reaction in DMSO. All these conjugates proved immunogenic in mice, which means the suitability of alternative (alternative) conjugation chemistry in addition to periodate oxidation/reductive amination chemistry, and alternative (alternative) conjugate properties (such as SPR).
TABLE 15 Process conditions and alternative chemical selection for GBS serotype II-CRM varying periodate Oxidation/reductive amination chemistry197Effect of the conjugate
Figure BDA0003350538080001031
GBS serotype III polysaccharide-CRM197Conjugates
Conjugates generated using PO/RAC and activated polysaccharide with DO between 10 and 17 (about 6 to 10% sugar epitope modification) in DMSO were shown to be immunogenic in mice (conjugates 25 and 30). Conjugates with a DO of 2.9 (about 34% saccharide epitope modification) or with a high saccharide/protein ratio (2.1) (conjugates 26 and 27, respectively) showed relatively low immunogenicity. The conjugate generated using PO/RAC and a polysaccharide with a sialylation level of 81% (19% desialylated) showed immunogenicity (conjugate 30). However, the conjugate produced using a polysaccharide with a sialylation degree of 58% (42% desialylation) showed poor immunogenicity (conjugate 29). When the properties of all other conjugate molecules are similar ( conjugates 25 and 28, respectively), the conjugates generated by PO/RAC in DMSO are somewhat more immunogenic than the conjugates generated by PO/RAC in aqueous media. The results are shown in Table 16.
TABLE 16 Process conditions for varying periodate Oxidation/reductive amination chemistry to GBS serotype III-CRM 197Effect of the conjugate
Figure BDA0003350538080001032
Figure BDA0003350538080001041
Additional conjugate variants were generated using alternative conjugation chemistries and conjugation molecule properties (results are shown in table 17). Conjugates 31 to 35 were generated by PO/RAC and deliberately varying the conjugation parameters to produce conjugates with different MW. Conjugate 36 is generated by reacting a polysaccharide with carbonyl bis-triazole (CDT) and performing a conjugation reaction in DMSO. Conjugate 37 was generated by oxidation of the polysaccharide using TEMPO reagent (instead of sodium periodate) followed by conjugation in DMSO using reductive amination chemistry, as detailed in example 3 above. All these conjugates proved to be immunogenic in mice, which represents an alternative conjugation chemistry in addition to periodate oxidation/reductive amination chemistry and the suitability (suivability) of alternative conjugate properties such as MW. Conjugates generated using as low as 5 DO (about 20% saccharide epitope modification) were still immunogenic in mice (conjugate 32 in table 17) compared to conjugates generated with 2.9 DO (about 34% saccharide epitope modification) (conjugate 26 in table 16 above).
TABLE 17 Process conditions and alternative chemical selection for GBS serotype III-CRM varying periodate Oxidation/reductive amination chemistry197Effect of the conjugate
Figure BDA0003350538080001042
Figure BDA0003350538080001051
GBS serotype IV polysaccharide-CRM197Conjugates
Conjugates generated using activated polysaccharides (activated polysaccharides) with PO/RAC and its DO between 6.9 and 14.2 (about 7 to 14% carbohydrate epitope modification) show immunogenicity in mice (conjugates 38 to 41). Conjugates generated using PO/AC and its polysaccharide with a sialylation degree of 60% (40% desialylation) also showed immunogenicity (conjugate 41). The results are shown in table 18.
TABLE 18 Process conditions for varying periodate Oxidation/reductive amination chemistry to GBS serotype IV-CRM197Effect of the conjugate
Figure BDA0003350538080001052
Figure BDA0003350538080001061
Alternative conjugation chemistries and conjugate molecular properties were used to generate additional conjugate variants (conjugate variants). The results are shown in table 19. Conjugates 42 and 45 were generated by PO/RAC and deliberately varying the conjugation parameters to produce conjugates with high DO (lower oxidation level) and high SPR, respectively. Conjugate 43 is generated by reacting the polysaccharide with carbonyl bis-triazole (CDT) and performing the conjugation reaction in DMSO. Conjugate 44 was generated by oxidation of the polysaccharide using TEMPO reagent (instead of sodium periodate) followed by conjugation in DMSO using reductive amination chemistry, as detailed in example 3 above. All serotype IV conjugates were shown to be immunogenic in mice, indicating the suitability of alternative conjugation chemistries in addition to periodate oxidation/reductive amination chemistry, as well as conjugate properties such as SPR. Conjugates generated with a DO (lower oxidation) of up to at least 20 (about 5% saccharide epitope modification) remain immunogenic in mice.
TABLE 19 Process conditions and alternative chemical selection for GBS serotype IV-CRM varying periodate Oxidation/reductive amination chemistry197Effect of the conjugate
Figure BDA0003350538080001062
Figure BDA0003350538080001071
GBS serotype V polysaccharide-CRM197Conjugates
Conjugates generated using PO/RAC and its activated polysaccharide with a DO of 4.4 to 14.6 (about 7 to 23% saccharide epitope modification) were shown to be immunogenic in mice (conjugates 46 and 47). The desialylated (5% sialylation degree) conjugate was not immunogenic (conjugate 49), whereas the conjugate generated using the PO/RAC method using an aqueous solvent gave a weak immune response (conjugate 48). The results are shown in table 20.
Table 20.Method conditions for changing periodate oxidation/reductive amination chemistry to GBS serotype V-CRM197Effect of the conjugate
Figure BDA0003350538080001072
Alternative conjugation chemistries and conjugation molecule properties were used to generate additional conjugate variants. The results are shown in table 21. Conjugates 50 and 53 were generated by PO/RAC and deliberately varying the conjugation parameters to produce conjugates with lower degrees of sialylation (81% sialylation) and low Molecular Weight (MW), respectively. Conjugate 51 was generated by reacting the polysaccharide with carbonyl bis-triazole (CDT) and performing the conjugation reaction in DMSO. Conjugate 52 was generated by oxidation of the polysaccharide using TEMPO reagent (instead of sodium periodate) followed by conjugation in DMSO using reductive amination chemistry, as detailed in example 3 above. All serotype V conjugates (except those generated using CDT chemistry) were shown to be immunogenic in mice, indicating the suitability of alternative conjugation chemistries in addition to periodate oxidation/reductive amination chemistry, as well as conjugate properties such as MW. Conjugates formed using CDT chemistry showed significantly less immunogenicity compared to other conjugates formed by RAC. The immune response provided by the 81% sialylated conjugate (conjugate 50) was weaker compared to the > 95% sialylated conjugate (conjugate 53), but stronger than that of the 5% sialylated conjugate (conjugate 49 in table 20 above).
TABLE 21 Process conditions and alternative chemical selection for GBS serotype V-CRM varying periodate Oxidation/reductive amination chemistry197Effect of the conjugate
Figure BDA0003350538080001081
Example 5: GBS III-CRM197And GBS V-CRM197Monovalent conjugate vaccines produce OPA responses in mice
1mcg, 0.1mcg or 0.01mcg with subcutaneous route at weeks 0, 3 and 6CRM197Conjugated Group B Streptococcus (GBS) serotype III (GBS III-CRM)197) Or with CRM197Conjugated GBS serotype V (GBS V-CRM)197) Female CD-1 mice were immunized (imminize) three times. Post-dose (Post dose three) (PD3) sera were evaluated by opsonophagocytic assay (OPA). OPA was performed as described in example 4. Both conjugates induced OPA responses in mice (table 22). Samples with no detectable OPA reaction were assigned a value of 50.
TABLE 22 GBS III and GBS V conjugates induce OPA responses in mice
Figure BDA0003350538080001091
Example 6: GBS Ia-CRM197、GBS Ib-CRM197、GBSⅡ-CRM197、GBSⅢ-CRM197、GBS IV-CRM197And GBS V-CRM197Monovalent conjugate vaccines produce OPA responses in mice
At weeks 0, 3 and 6 via subcutaneous route to contain 1mcg with CRM197Vaccines of conjugated individual GBS Capsular Polysaccharide (CP) were immunized three times to six groups of female CD-1 mice. Preliminary studies showed that mice did not have pre-existing OPA titers (pre-existing OPA titer) for any of the six serotypes tested. Sera from PD3 were analyzed by OPA against the homologous (cognate) GBS serotype contained in the vaccine. OPA was performed as described in example 4. The results are shown in tables 23 and 24 below.
TABLE 23 CPS-CRM with individual GBS197Geometric mean OPA titers in mice following conjugate immunization
Figure BDA0003350538080001101
TABLE 24 CPS-CRM with individual GBS197Fold increase in OPA titre in mice following conjugate immunization
Figure BDA0003350538080001102
Note that the fold-up was calculated assuming that the mice did not have pre-existing titers.
Example 7: and from GBS Ia-CRM197、GBS Ib-CRM197、GBSⅡ-CRM197、GBSⅢ-CRM197、GBS IV-CRM197And GBS V-CRM197IgG-compared serum opsonin Activity of mice immunized with monovalent conjugate vaccines
1mcg of CRM at weeks 0, 3 and 6 via the subcutaneous route197Conjugated individual GBS CPs female CD-1 or BALB/c mice were immunized three times. Using AlPO4Or QS-21 as an adjuvant. PD3 serum was tested by serotype specific OPA (serotype-specific OPA), followed by isolation of immunoglobulin G fraction (immunoglobulin G fraction) and testing for OPA activity. Purified IgG OPA activity was normalized to 5mg/ml (within the range of IgG amount in normal mouse serum). All six GBS CPS conjugates elicit IgG antibodies with opsonin activity (fig. 1).
Example 8: monovalent conjugate vaccines of GBS Ia-TT, GBS Ib-TT, GBS II-TT, GBS III-TT, GBS IV-TT and GBS V-TT produce OPA responses in rabbits
Rabbits were immunized three times with 50mcg/ml of GBS serotype la polysaccharide conjugated to tetanus toxoid, 10mcg/ml of GBS serotype lb polysaccharide conjugated to tetanus toxoid, 50mcg/ml of GBS serotype ii polysaccharide conjugated to tetanus toxoid, 50mcg/ml of GBS serotype iii polysaccharide conjugated to tetanus toxoid, 50mcg/ml of GBS serotype IV polysaccharide conjugated to tetanus toxoid, or 50mcg/ml of GBS serotype V polysaccharide conjugated to tetanus toxoid, wherein Complete Freund's Adjuvant was combined in the first dose and Incomplete Freund's Adjuvant was combined in the second and third doses. The conjugate used polysaccharides with sialic acid levels > 95% and CDAP (1-cyano-4-dimethylaminopyridine tetrafluoroborate manufactured chemically, PD3 immunoreactivity measured by OPA as described in example 4 serum titers are shown in table 25, while purified IgG titers are shown in table 26 below GBS serotypes Ia, Ib, ii, iii, IV and V polysaccharides conjugated to TT are highly immunogenic in rabbits.
TABLE 25 geometric mean OPA titres of rabbit sera after immunization with Individual GBS CPS-TT conjugates
Figure BDA0003350538080001111
TABLE 26 geometric mean OPA titres of rabbit sera after immunization with Individual GBS CPS-TT conjugates
Figure BDA0003350538080001121
Example 9: hexavalent GBS conjugate vaccines produce OPA responses in non-human primates
Three groups of rhesus monkeys (rhesus macaque) were immunized three times via intramuscular route (intramulcular) with hexavalent (hexavalent) group B streptococcus (GBS6) vaccine at weeks 0, 4 and 8. GBS6 vaccines include GBS Ia-CRM197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197. Two groups include aluminum phosphate (AlPO)4) As adjuvant and either 5mcg of each conjugate or 50mcg of each conjugate was administered. The third group was administered 5mcg of each conjugate and no adjuvant. The following table 27 describes an immunization schedule (immunization schedule).
TABLE 27 Immunization schedule of rhesus monkeys of rhesus macaques
Figure BDA0003350538080001122
Figure BDA0003350538080001131
All six GBS serotypes contained in the vaccine were analyzed by OPA in preimmune serum (preimmune serum) and in serum from PD 3. OPA was performed as described in example 4. The results are shown in tables 28 and 29 below. In all six serotypes, aluminum phosphate (AlPO) was added4) Adjuvant formulations caused detectable OPA responses (increase in pre-immune (pre) to PD3 titers; or fold-up pre-immune/PD 3(pre/PD3) >1). A 5 mcg/conjugate dose without adjuvant elicited a detectable OPA response to 5 of the 6 serotypes.
TABLE 28 geometric mean OPA titres of rhesus monkeys before and after immunization with GBS6
Figure BDA0003350538080001132
TABLE 29 fold increase in OPA titer in rhesus monkeys after immunization with GBS6
Figure BDA0003350538080001133
Example 10: hexavalent GBS conjugate vaccines produce OPA responses in rats
Polysaccharide conjugate vaccine in GBS6 (formulated as in example 9, with or without aluminium phosphate (AlPO) at 5mcg/ml via subcutaneous route (subnutaneousy)4) For example) female history-Dawley rats (Sprague-Dawley rat) were immunized twice. Both pre-immune (preimmune) (baseline) and post-dose (post-dose) sera were evaluated for OPA assay titers (OPA assay titer) against all six homologous (cognate) GBS serotypes. OPA titers were measured for each serotype in a GBS 384well assay format (GBS 384well assay format) and fold-up (fold rise) was calculated. Rats administered with GBS6 vaccine had a robust functional antibody response (functional antibody response) to each serotype after the second dose; when there is no AlPO4When present, a 7 to 205 fold increase was seen between serotypes, whereas AlPO was present 4This range is 11 to 294 fold (table 30). TABLE 30 Titer fold increase after dose 2 (PD2) in the opsonophagocytosis Activity (OPA) assay in rats immunized with hexavalent GBS conjugate vaccine
Figure BDA0003350538080001141
Example 11: pregnant females immunized with monovalent or hexavalent GBS glycoconjugate vaccines exhibit protective effects against GBS III or V infection after birth
By subcutaneous route to a conjugate containing 5mcg/ml of each conjugate and 100mcg/ml of AlPO as described in example 94GBS6 vaccine, GBS III or V monovalent saccharide conjugate vaccine (monovalent glycoconjugate vaccine) (each containing 10mcg/ml of conjugate and 100mcg/ml of AlPO4) Female CD-1 mice were immunized three times with, or with a vehicle control alone (vehicle control alone). Mice were gestated (bred) prior to the third immunization. Progeny of immunized mice were challenged (challenge) with a lethal dose (lethase) of GBS serotype iii or GBS serotype V bacteria, depending on the vaccine received, and survival was monitored for 90 hours. With GBS6+ AlPO4Or GBS III-CRM197+AlPO4The immunized female parent (dam) provided significant protection to its pups (p)<0.0001) against lethal GBS serotype iii challenge. Similarly, with GBS V-CRM 197+AlPO4The immunized female parent (dam) provided significant protection to its pups (p)<0.0001) against lethal GBS serotype V challenge (lethal GBS serotype V challenge). The results are shown in table 31.
TABLE 31 immunization with monovalent and hexavalent GBS vaccines increases survival of offspring
Figure BDA0003350538080001151
Example 12: passive Immunization (Passive Immunization) of the GBS III monoclonal antibody in young mice shows a protective effect
Mice were immunized with a pentavalent vaccine comprising serotypes Ia, Ib, II, III and V to generate group B streptococcus serotype III (gbs III) monoclonal antibodies (mabs). GBS III-specific mAb clones (GBS III-specific mAb clone) were selected and mAbs recognizing the CP of GBS III were generated using standard procedures. GBS serotype III mAb was passively administered to young mice 16 hours prior to challenge with clinical GBS III isolates (10 for each group; 2 independent experiments are shown). Blood was collected four hours after challenge and the remaining cfu (remaining cfu) was calculated. Treatment with GBS III mAb reduced cfu recovery in young mice by 4log or more (see table 32).
TABLE 32 GBS III mAb reduction of CFU recovery in pups
Figure BDA0003350538080001152
Figure BDA0003350538080001161
Example 13: passive immunization of GBS Ib, III and V monoclonal antibodies in pregnant mice showed protective effects on postnatal offspring thereof
The vaccine was prepared from a pentavalent vaccine (GBS Ia-CRM) using standard procedures197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197And GBS V-CRM197) Immunized mice produce monoclonal antibodies (mabs). The mAb was then confirmed to specifically recognize (specific recogniting) the capsular polysaccharides of each of the 5 serotypes. About 24 to 48 hours before delivery of the pregnant mice, a 500mcg/ml dose of GBS serotype ib (GBS ib) mAb, GBS serotype iii (GBS iii) mAb, GBS serotype v (GBS v) mAb, or isotype-matched (isotype-matched) control mAb was passively administered to the pregnant mice. The offspring of immunized female mice are challenged with a lethal dose of GBS Ib, GBS III or GBS V bacteria 24 to 48 hours after birth. Survival was monitored for 96 hours. The survival of female born pups immunized with GBS Ib, GBS III, or GBS V mAb was significantly higher after GBS challenge compared to those immunized with control mAb (see below)Table 33).
TABLE 33 GBS III & V mAb increase survival of offspring
Figure BDA0003350538080001162
Example 14: stability of GBS conjugates
GBS Ia-CRM was formulated separately at different pH levels in 10mM succinate-phosphate and 155mM NaCl197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197To test the stability of the conjugates under accelerated storage conditions. The percent change in molecular weight (determined by SEC MALLS) was measured after 4 weeks storage at 50 ℃. The results are shown in fig. 2 to 7.
GBS Ia-CRM was tested under various buffer conditions shown in Table 34197、GBS Ib-CRM197、GBS III-CRM197And GBS IV-CRM197Sialylation stability of the conjugate. After 1 month of storage at 37 ℃, free sialic acid (N-acetylneuraminic acid); NANA) was measured using HPLC. The results are shown in FIG. 8.
Two studies have shown that the conjugate performs better above pH 6.0, and most ideally at about pH 6.5.
TABLE 34 buffer conditions for sialylation stability test
Figure BDA0003350538080001171
Example 15: GBS6 formulations
To determine the choice of buffer, GBS Ia-CRM was formulated together using the same buffer conditions as shown in Table 33 above197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197(GBS 6). The actual pH of such formulations was tested at the following time points: 0 (at the time of formulation manufacture), at 5 DEG CAfter 1 month, after 1 month at 25 ℃ and after 1 month at 37 ℃. A pH shift was seen in the formulation using amber salt as buffer, whereas no shift was seen in the formulation using histidine as buffer. The results are shown in fig. 9 to 10.
The effect of histidine buffer concentration on the binding of GBS conjugates to aluminum was also tested. The conjugate (10mcg/ml and 40mg/ml of each serotype) at two different concentrations and several histidine tests at different concentrations included 150mM NaCl, 0.01% polysorbate-80 at pH 6.5 and 0.5mg/ml as AlPO 4The formulation of aluminum of (1). The percentage of conjugates bound to aluminum was determined by measuring the total amount of each conjugate and the amount of each conjugate bound to aluminum in the vaccine. Measuring the bound conjugate by: the vaccine formulations were centrifuged, the aluminum pellet resuspended, the aluminum dissolved and the bound conjugate measured using turbidimetry (nephelometry) with serotype-specific polyclonal antibodies (serotype-specific polyclonal antibodies) against each serotype. The results are shown in fig. 11 to 12. The concentration of histidine buffer was found to affect the percentage of aluminum binding of each serotype, with the effect being more pronounced at lower doses than at higher doses.
An agitation study (agitation study) was performed to determine the amount of polysorbate-80 (PS80) that was desired. The assay contained 20mM histidine, 150mM NaCl, 0.5mg/ml AlPO4(when present), and the percentage of total antigenic loss (total antigenic lost) of GBS6 formulations without PS80, with 0.01% PS80, 0.02% PS80, or 0.03% PS80(pH 6.5) under stirring pressure (agitation stress). The syringe (syringe) pre-filled with formulation was stirred at 500RPM for 72 hours at room temperature. Control samples (without stirring) were stored at room temperature for 72 hours. The results are shown in fig. 13.
The aluminum concentration in GBS6 formulations was also investigated to determine its effect on binding of GBS conjugates to aluminum. For aluminum phosphate (AlPO) containing 10mM histidine, 150mM NaCl, 0.02% PS80, and 0.25mg/ml, 0.5mg/ml, or 0.75mg/ml, or 1.0mg/ml4) GBS6 formulation of aluminum (pH 6.5) the percentage of conjugate bound to aluminum was tested. And AlPO4Percent incorporation byTo AlPO4The concentration increases. The results are shown in fig. 14.
Example 16: GBS6 freeze-dried formulation
Test GBS6(GBS Ia-CRM)197、GBS Ib-CRM197、GBS II-CRM197、GBS III-CRM197、GBS IV-CRM197And GBS V-CRM197) Stability of various freeze-dried formulations of (a). Low (10mcg/ml) and high (50mcg/ml) dose formulations comprising 20mM histidine (pH 6.5), 0.02% PS80, about 28mM NaCl, and 5.5%, 7.0%, or 8.5% (w/v) sucrose were freeze-dried. The stability of the freeze-dried formulations was tested by measuring pH and moisture after 4 months at 5 ℃, 4 months at 37 ℃ and 1 month at 50 ℃. All formulations were stable according to pH and moisture (data not shown). Furthermore, all formulations were tested for the percentage of antigenic recovery (antigen recovery) of each serotype after 1, 4 and 9 months at 5 ℃ and 37 ℃ and after 1, 2 and 4 weeks at 50 ℃. The results are shown in fig. 15 to 20.
The following variations of excipients for a 40mcg/ml dose of GBS6 formulation were also prepared and evaluated: 1) 7% (w/v) sucrose, 2) 2% (w/v) sucrose and 4% (w/v) mannitol, 3) 3% (w/v) sucrose and 3% (w/v) mannitol, 4) 2% (w/v) sucrose and 4% (w/v) glycine, or 5) 3% (w/v) sucrose and 3% (w/v) glycine. All five formulations were stable in pH and moisture (moisture) after 3 months at 5 deg.C, 3 months at 25 deg.C, 3 months at 37 deg.C and 1 month at 50 deg.C (data not shown). In addition, all formulations were tested for percent antigenic recovery of each serotype after 1, 3 and 7 months at 52-8 ℃, 25 ℃ and 37 ℃, and after 1, 2 and 4 weeks at 50 ℃. The results are shown in fig. 21 to 25.
The percentage of antigen bound to aluminium phosphate adjuvant of GBS6 vaccine in reconstituted freeze-dried formulations (reconstituted lyophilised formulations) and liquid formulations (liquid formulations) was tested using turbidimetry. Lyophilized and liquid formulations containing 20mM histidine, 0.02% PS80, 7.0% (w/v) sucrose and 500mcg/ml aluminum as aluminum phosphate were prepared in both low (10mcg/ml) and high (50mcg/ml) doses. Altered sodium chloride (NaCl) concentrations were also tested to determine the effect on antigen binding. The results for the freeze-dried and liquid formulations are shown in tables 35 and 36, respectively. Low dose formulations of both freeze-dried and liquid compositions have comparable results to each other when using NaCl concentrations of about 150mM or higher.
TABLE 35 percentage of antigen bound to aluminum phosphate in formulated freeze-dried formulations with varying amounts of NaCl
Figure BDA0003350538080001191
Figure BDA0003350538080001201
TABLE 36 percentage of antigen bound to aluminum phosphate in liquid formulations with varying amounts of NaCl
Figure BDA0003350538080001202
Aspects of the invention
The following items describe further embodiments of the invention:
C1. an immunogenic polysaccharide-protein conjugate comprising a Group B Streptococcus (GBS) capsular polysaccharide and a carrier protein, wherein the capsular polysaccharide has a sialic acid level of greater than about 60%.
C2. The immunogenic conjugate of C1, wherein the capsular polysaccharide is selected from the group consisting of: serotypes Ia, Ib, II, III, IV, V, VI, VII, VIII and IX.
C3. An immunogenic conjugate according to C2, wherein the capsular polysaccharide is serotype Ia.
C4. The immunogenic conjugate of C2, wherein the capsular polysaccharide is serotype Ib.
C5. The immunogenic conjugate of C2, wherein the capsular polysaccharide is serotype II.
C6. The immunogenic conjugate of C2, wherein the capsular polysaccharide is serotype III.
C7. The immunogenic conjugate of C2, wherein the capsular polysaccharide is serotype IV.
C8. The immunogenic conjugate of C2, wherein the capsular polysaccharide is serotype V.
C9. The immunogenic conjugate of C2, wherein the capsular polysaccharide is serotype VI.
C10. The immunogenic conjugate of C2, wherein the capsular polysaccharide is serotype VII.
C11. The immunogenic conjugate of C2, wherein the capsular polysaccharide is serotype VIII.
C12. The immunogenic conjugate of C2, wherein the capsular polysaccharide is serotype IX.
C13. The immunogenic conjugate of any one of C1-C12, wherein the sialic acid level of the capsular polysaccharide is greater than about 95%.
C14. The immunogenic conjugate of any one of C1-C13, wherein the sialic acid level of the capsular polysaccharide is about 100%.
C15. The immunogenic conjugate of any one of C1-C14, wherein the capsular polysaccharide has at least about 0.6mM sialic acid per mM polysaccharide.
C16. The immunogenic conjugate of any one of C1-C15, wherein the capsular polysaccharide has at least about 0.65mM sialic acid per mM polysaccharide.
C17. The immunogenic conjugate of any one of C1-C16, wherein the capsular polysaccharide has at least about 0.7mM sialic acid per mM polysaccharide.
C18. The immunogenic conjugate of any one of C1-C17, wherein the capsular polysaccharide has at least about 0.75mM sialic acid per mM polysaccharide.
C19. The immunogenic conjugate of any one of C1-C18, wherein the capsular polysaccharide has at least about 0.8mM sialic acid per mM polysaccharide.
C20. The immunogenic conjugate of any one of C1-C19, wherein the capsular polysaccharide has at least about 0.85mM sialic acid per mM polysaccharide.
C21. The immunogenic conjugate of any one of C1-C20, wherein the capsular polysaccharide has at least about 0.9mM sialic acid per mM polysaccharide.
C22. The immunogenic conjugate of any one of C1-C21, wherein the capsular polysaccharide has at least about 0.95mM sialic acid per mM polysaccharide.
C23. The immunogenic conjugate of any one of C1-C22, wherein the capsular polysaccharide has a molecular weight of about 5kDa to about 1,000 kDa.
C24. The immunogenic conjugate of any one of C1-C23, wherein the capsular polysaccharide has a molecular weight of about 25kDa to about 750 kDa.
C25. The immunogenic conjugate of any one of C1-C24, wherein the capsular polysaccharide has a molecular weight of about 25kDa to about 400 kDa.
C26. The immunogenic conjugate of any one of C1-C25, wherein the capsular polysaccharide has a molecular weight of about 25kDa to about 200 kDa.
C27. The immunogenic conjugate of any one of C1-C25, wherein the capsular polysaccharide has a molecular weight of about 100kDa to about 400 kDa.
C28. The immunogenic conjugate of any one of C1 to C27, wherein the conjugate has a molecular weight of about 300kDa to about 20,000 kDa.
C29. The immunogenic conjugate of any one of C1 to C28, wherein the molecular weight of the conjugate is about 1,000kDa to about 15,000 kDa.
C30. The immunogenic conjugate of any one of C1 to C29, wherein the molecular weight of the conjugate is about 1,000kDa to about 10,000 kDa.
C31. The immunogenic conjugate of any one of C1-C30, wherein the capsular polysaccharide is about 0% to about 40% O-acetylated.
C32. The immunogenic conjugate of any one of C1-C31, wherein the capsular polysaccharide is less than about 5% O-acetylated.
C33. The immunogenic conjugate of any one of C1-C32, wherein the capsular polysaccharide is less than about 4% O-acetylated.
C34. The immunogenic conjugate of any one of C1-C33, wherein the capsular polysaccharide is less than about 3% O-acetylated.
C35. The immunogenic conjugate of any one of C1-C34, wherein the capsular polysaccharide is less than about 2% O-acetylated.
C36. The immunogenic conjugate of any one of C1-C35, wherein the capsular polysaccharide is less than about 1% O-acetylated.
C37. The immunogenic conjugate of any one of C1-C36, wherein the capsular polysaccharide comprises at least about 0.1mM O-acetate per mM saccharide repeat unit.
C38. The immunogenic conjugate of any one of C1-C37, wherein the capsular polysaccharide comprises at least about 0.2mM O-acetate per mM saccharide repeat unit.
C39. The immunogenic conjugate of any one of C1-C38, wherein the capsular polysaccharide comprises at least about 0.3mM O-acetate per mM saccharide repeat unit.
C40. The immunogenic conjugate of any one of C1-C39, wherein the capsular polysaccharide comprises at least about 0.35mM O-acetate per mM saccharide repeat unit.
C41. The immunogenic conjugate of any one of C1-C40, wherein the capsular polysaccharide comprises about 0.4mM O-acetate per mM saccharide repeat unit.
C42. The immunogenic conjugate of any one of C1-C41, wherein the capsular polysaccharide comprises less than about 0.01mM O-acetate per mM saccharide repeat unit.
C43. The immunogenic conjugate of any one of C1-C42, wherein the capsular polysaccharide comprises less than about 0.05mM O-acetate per mM saccharide repeat unit.
C44. The immunogenic conjugate of any one of C1-C43, wherein the capsular polysaccharide comprises less than about 0.04mM O-acetate per mM saccharide repeat unit.
C45. The immunogenic conjugate of any one of C1-C44, wherein the capsular polysaccharide comprises less than about 0.03mM O-acetate per mM saccharide repeat unit.
C46. The immunogenic conjugate of any one of C1-C45, wherein the capsular polysaccharide comprises less than about 0.02mM O-acetate per mM saccharide repeat unit.
C47. The immunogenic conjugate of any one of C1 to C46, wherein the polysaccharides are each independently conjugated to a carrier protein.
C48. The immunogenic conjugate of any one of C1-C47, wherein the carrier protein is CRM197Or tetanus toxoid.
C49. The immunogenic conjugate of any one of C1-C48, wherein the carrier protein is CRM197
C50. A process for isolating capsular polysaccharides comprising reacting an organic reagent with a cell broth (cell broth) comprising a capsular polysaccharide producing bacterium.
C51. The method of C50, wherein the bacterium is not lysed (not lysed).
C52. The method according to C50 or 51, wherein the bacterium is heat killed.
C53. The method according to any one of C50 to 52, wherein the method further comprises a centrifugation step to provide a cell paste (cell paste).
C54. The method of any one of C50 to 53, wherein the method further comprises a filtration step.
C55. The method of C54, wherein the filtration step is diafiltration (diafiltration).
C56. The process of any one of C50 to 55, wherein the capsular polysaccharide producing bacterium is selected from the group consisting of: streptococcus agalactiae (Streptococcus agalactiae), Streptococcus pneumoniae (Streptococcus pneumoniae), Staphylococcus aureus (Staphylococcus aureus), Neisseria meningitidis (Neisseria meningitidis), Escherichia coli (Escherichia coli), Salmonella typhi (Salmonella typhi), Haemophilus influenzae (Haemophilus influenzae), Klebsiella pneumoniae (Klebsiella pneumoniae), Enterococcus faecium (Enterococcus faecalis), and Enterococcus faecalis (Enterococcus faecalis).
C57. The method of C56, wherein the bacterium is Streptococcus agalactiae.
C58. The method according to any one of C50 to 57, wherein the organic reagent is a derivatized hydroxylamine compound (derivatized hydroxyl amine compound).
C59. The method of any one of C50 to 58, wherein the hydroxylamine is any of the hydroxylamines listed in table 2 of example 2.
C60. The method of any one of C50 to 59, wherein the hydroxylamine is selected from the group consisting of: dibenzylhydroxylamine (dibenzylhydroxylamine); diethylhydroxylamine (diethyl hydroxylamine); hydroxylamine (hydroxyimine); ethylenediamine (ethylenediamine); triethylenetetramine (triethylenetramine); 1,1,4,7,10,10 hexamethyltriethylenetetramine (1,1,4,7,10,10 hexamethylene triene tetramine); and 2,6,10, Trimethyl 2,6,10triazaundecane (2,6,10, trimethy 2,6,10 triazaundecane).
C61. The method of any one of C50 to C60, wherein the concentration of hydroxylamine is about 5mM to about 200 mM.
C62. The method of any one of C50 to C61, wherein the pH of the reaction is about 5.5 to about 9.5.
C63. The method of any one of C50 to C62, wherein the reaction is carried out at a temperature of about 20 ℃ to about 85 ℃.
C64. The method of any one of C50 to C63, wherein the reaction time is about 10 hours to about 90 hours.
C65. A process for manufacturing an immunogenic polysaccharide-protein conjugate according to any one of C1 to C49, wherein the capsular polysaccharide is isolated according to the process of any one of C50 to C64.
C66. An immunogenic polysaccharide-protein conjugate comprising a capsular polysaccharide manufactured by a process as in any one of C50 to C64.
C67. An immunogenic composition comprising an immunogenic polysaccharide-protein conjugate as in any one of C1 to C49 or C66.
C68. An immunogenic composition comprising a polysaccharide-protein conjugate, wherein the conjugate comprises a capsular polysaccharide from Group B Streptococcus (GBS) serotype IV and at least one additional serotype selected from the group consisting of: ia. Ib, II, III, V, VI, VII, VIII and IX.
C69. The immunogenic composition of C68 wherein the at least one additional serotype is Ia.
C70. The immunogenic composition of C69, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype Ib.
C71. An immunogenic composition according to C69 or C70, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype II.
C72. The immunogenic composition of any one of C69-C71, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype III.
C73. The immunogenic composition of any one of C69-C72, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype V.
C74. The immunogenic composition of any one of C69-C73, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VI.
C75. The immunogenic composition of any one of C69-C74, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VII.
C76. The immunogenic composition of any one of C69-C75, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VIII.
C77. The immunogenic composition of any one of C69-C76, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype IX.
C78. The immunogenic composition of C68 wherein the at least one additional serotype is Ib.
C79. The immunogenic composition of C78, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype II.
C80. An immunogenic composition according to C78 or C79, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype III.
C81. The immunogenic composition of any one of C78-C80, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype V.
C82. The immunogenic composition of any one of C78-C81, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VI.
C83. The immunogenic composition of any one of C78-C82, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VII.
C84. The immunogenic composition of any one of C78-C83, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VIII.
C85. The immunogenic composition of any one of C78-C84, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype IX.
C86. The immunogenic composition of C68 wherein the at least one additional serotype is II.
C87. The immunogenic composition of C86, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype III.
C88. An immunogenic composition according to C86 or C87, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype V.
C89. The immunogenic composition of any one of C86-C88, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VI.
C90. The immunogenic composition of any one of C86-C89, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VII.
C91. The immunogenic composition of any one of C86-C90, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VIII.
C92. The immunogenic composition of any one of C86-C91, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype IX.
C93. The immunogenic composition of C68 wherein the at least one additional serotype is III.
C94. The immunogenic composition of C93, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype V.
C95. The immunogenic composition of C93 or C94, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VI.
C96. The immunogenic composition of any one of C93-C95, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VII.
C97. The immunogenic composition of any one of C93-C96, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VIII.
C98. The immunogenic composition of any one of C93-C97, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype IX.
C99. The immunogenic composition of C68 wherein the at least one additional serotype is V.
C100. The immunogenic composition of C99, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VI.
C101. The immunogenic composition of any one of C99 or C100, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VII.
C102. The immunogenic composition of any one of C99-C101, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VIII.
C103. The immunogenic composition of any one of C99-C102, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype IX.
C104. The immunogenic composition of C68 wherein the at least one additional serotype is VI.
C105. The immunogenic composition of claim 104, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VII.
C106. The immunogenic composition of any one of C104 or C105, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VIII.
C107. The immunogenic composition of any one of C104-C106, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype IX.
C108. The immunogenic composition of C68, wherein the at least one additional serotype is VII.
C109. The immunogenic composition of C108, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype VIII.
An immunogenic composition of any one of C110 or C109, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype IX.
C111. The immunogenic composition of C68 wherein the at least one additional serotype is VIII.
C112. The immunogenic composition of C111, wherein the composition further comprises a conjugate comprising a capsular polysaccharide from GBS serotype IX.
C113. The immunogenic composition of C112, wherein the at least one additional serotype is IX.
C114. An immunogenic composition comprising a polysaccharide-protein conjugate, wherein the conjugate comprises capsular polysaccharides from GBS serotypes Ia, Ib, II, III and IV.
C115. An immunogenic composition comprising polysaccharide-protein conjugates, wherein the conjugates comprise capsular polysaccharides from GBS serotypes Ia, Ib, II, III and V.
C116. An immunogenic composition comprising a polysaccharide-protein conjugate, wherein the conjugate comprises capsular polysaccharides from GBS serotypes Ia, Ib, II, III, IV and V.
C117. An immunogenic composition comprising a polysaccharide-protein conjugate comprising at least four GBS capsular polysaccharide serotypes selected from the group consisting of: ia. Ib, II, III, IV, V, VI, VII, VIII and IX.
C118. The immunogenic composition of C117, wherein the composition comprises at least five GBS capsular polysaccharide serotypes.
C119. An immunogenic composition according to C117 or C118, wherein the composition comprises at least six GBS capsular polysaccharide serotypes.
C120. The immunogenic composition of any one of C117 to C119, wherein the composition comprises at least seven GBS capsular polysaccharide serotypes.
C121. The immunogenic composition of any one of C117 to C120, wherein the composition comprises at least eight GBS capsular polysaccharide serotypes.
C122. The immunogenic composition of any one of C117 to C121, wherein the composition comprises at least nine GBS capsular polysaccharide serotypes.
C123. The immunogenic composition of any one of C117 to C122, wherein the composition comprises GBS capsular polysaccharide serotype V.
C124. The immunogenic composition of any one of C117 to C123, wherein the composition does not have immune interference (immuneinterference).
C125. The immunogenic composition of any one of C67-C124, wherein the composition further comprises a pharmaceutically acceptable excipient, buffer, stabilizer, adjuvant, cryoprotectant(s), salt(s), divalent cation(s), non-ionic detergent(s), free radical oxidation inhibitor(s), carrier(s), or mixture(s) thereof.
C126. The immunogenic composition of any one of C67-C125, wherein the composition further comprises a buffering agent.
C127. The immunogenic composition of C126, wherein the buffer is selected from the group consisting of: HEPES, PIPES, MES, Tris (tromethamine), phosphate, acetate, borate, citrate, glycine, histidine and succinate.
C128. The immunogenic composition of C127, wherein the buffering agent is histidine.
C129. The immunogenic composition of any one of C67-C128, wherein the composition further comprises a surfactant.
C130. The immunogenic composition of C129, wherein the surfactant is selected from the group consisting of: polyoxyethylene sorbitan fatty acid ester (polyoxyethylenesorbitan fatty acid ester), polysorbate-80, polysorbate-60, polysorbate-40, polysorbate-20 and polyoxyethylene alkyl ether (polyoxyethylenealkyl ether).
C131. The immunogenic composition of C130, wherein the surfactant is polysorbate-80.
C132. The immunogenic composition of any one of C67-C131, wherein the composition further comprises an excipient.
C133. The immunogenic composition of C132, wherein the excipient is selected from the group consisting of: starch, glucose, lactose, sucrose, trehalose (trehalose), raffinose, stachyose (stachyose), melezitose (melezitose), dextran (dextran), mannitol, lactitol (lactitol), palatinit (palatinit), gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, glycine, arginine, lysine, sodium chloride (NaCl), dried skim milk, glycerol, propylene glycol, water and ethanol.
C134. The immunogenic composition of C133, wherein the excipient is sodium chloride.
C135. The immunogenic composition of any one of C67-C134, wherein the composition further comprises an adjuvant.
C136. An immunogenic composition such as C135 wherein the adjuvant is an aluminium-based adjuvant or QS-21.
C137. The immunogenic composition of C136, wherein the aluminum-based adjuvant is selected from the group consisting of: aluminum phosphate, aluminum hydroxy phosphate, and aluminum hydroxide.
C138. The immunogenic composition of C137, wherein the adjuvant is aluminum phosphate.
C139. The immunogenic composition of C138, wherein the adjuvant is aluminum hydroxyphosphate.
C140. The immunogenic composition of any one of C67-C139, wherein the composition comprises a buffer, a surfactant, an excipient, and optionally an adjuvant, wherein the composition is buffered to a pH of about 6.0 to about 7.0.
C141. The immunogenic composition of any one of C67-C140, wherein the composition comprises histidine, polysorbate-80, NaCl, and optionally aluminum phosphate, wherein the composition is buffered to a pH of about 6.0 to about 7.0.
C142. The immunogenic composition of any one of C67-C141, wherein the composition comprises about 10mM to about 25mM histidine, about 0.01% to about 0.03% (v/w) polysorbate-80, about 10mM to about 250mM NaCl, and optionally about 0.25mg/ml to about 0.75mg/ml aluminum as aluminum phosphate.
C143. The immunogenic composition of any one of C67-C142, wherein the composition comprises a dose of about 5mcg/ml to about 50 mcg/ml.
C144. The immunogenic composition according to any one of C67 to C143, wherein the composition is lyophilized, optionally in the presence of at least one excipient.
C145. The immunogenic composition of C144, wherein the at least one excipient is selected from the group consisting of: starch, glucose, lactose, sucrose, trehalose, raffinose, stachyose, melezitose, dextran, mannitol, lactitol, arabitol, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, glycine, arginine, lysine, sodium chloride (NaCl), dried skim milk, glycerol, propylene glycol, water and ethanol.
C146. The immunogenic composition of C145, wherein the at least one excipient is sucrose.
C147. The immunogenic composition of any one of C144 to C146, wherein the composition comprises about 1% (w/v) to about 10% (w/v) of the at least one excipient.
C148. The immunogenic composition of any one of C144 to C147, wherein the composition comprises an additional excipient.
C149. The immunogenic composition of C148, wherein the additional excipient is mannitol or glycine.
C150. The immunogenic composition according to C148 or C149, wherein the composition comprises about 1% (w/v) to about 10% (w/v) additional excipient.
C151. The immunogenic composition of any one of C143 to C150, wherein the composition is reconstituted with water, water for injection (WFI), adjuvant suspension, or saline.
C152. An immunogenic composition as in any one of C67 to C151 for use as a medicament.
C153. The immunogenic composition of any one of C67-C152 for use in a method of eliciting an immune response to GBS in a subject.
C154. The immunogenic composition of C153, wherein the subject is a woman scheduled to become pregnant or a pregnant woman.
C155. The immunogenic composition of C154, wherein the female is in the second half of her pregnancy.
C156. The immunogenic composition of C155, wherein the pregnant female is at least 20 weeks gestation.
C157. The immunogenic composition of C156, wherein the pregnant female is between 27 and 36 weeks gestation.
C158. The immunogenic composition of C157, wherein the subject is an adult 50 years or older.
C159. The immunogenic composition of C158, wherein the subject is an adult aged 65 or older.
C160. The immunogenic composition of C159, wherein the subject is an adult of 85 years of age or older.
C161. The immunogenic composition of any one of C153 to C160, wherein the subject is immunocompromised.
C162. The immunogenic composition of C161, wherein the subject has a medical condition selected from the group consisting of: obesity, diabetes, HIV infection, cancer, cardiovascular disease or liver disease.
C163. The immunogenic composition of any one of C153-C162, wherein the group B streptococcus is streptococcus agalactiae.
C164. A method of eliciting an immune response to group B streptococcus comprising administering to a subject an effective amount of an immunogenic composition, such as any one of C67 to C150.
C165. A method of preventing or reducing a disease or condition associated with group B streptococcus in a subject, comprising administering to the subject an effective amount of an immunogenic composition, such as any one of 67 to C151.
C166. The method of C164 or C165, wherein the subject is a female intended to be pregnant or a pregnant female.
C167. The method of C166, wherein the female is in the second half of her pregnancy.
C168. Such as C166 or C167, wherein the pregnant female is at least 20 weeks gestation.
C169. The method of any one of C166-C168, wherein the pregnant female is 27 weeks to 36 weeks gestation.
C170. The method of C164 or C165, wherein the subject is an adult of 50 years or older.
C171. The method of C170, wherein the subject is an adult 65 years or older.
C172. The method of C170 or C171, wherein the subject is an adult of 85 years or older.
C173. The method of any one of C164 to C172, wherein the subject is immunocompromised.
C174. The method of C173, wherein the subject has a medical condition selected from the group consisting of: obesity, diabetes, HIV infection, cancer, cardiovascular disease, or liver disease.
C175. The method of any one of C164 to C174, wherein the group B streptococcus is Streptococcus agalactiae.
C176. An antibody that binds to a capsular polysaccharide in an immunogenic conjugate as in any one of C1 to C49 or C66.
C177. A composition comprising an antibody such as C176.
C178. A method of making an antibody comprising administering to a subject an immunogenic composition as in any of C67 to C151.
C179. An antibody produced by a method such as C178.
C180. A method of imparting passive immunity to a subject comprising the steps of:
(a) generating an antibody preparation using an immunogenic composition as in any of C67 to C151; and
(b) the antibody formulation is administered to a subject to confer passive immunity.
C181. A method of making an immunogenic polysaccharide-protein conjugate as any one of C1 to C49 or C66, comprising the steps of:
(a) reacting a GBS capsular polysaccharide with an oxidizing agent to produce an activated polysaccharide;
(b) reacting the activated polysaccharide with a carrier protein to produce a polysaccharide-protein conjugate.
C182. The process of C181, wherein step (b) is carried out in a polar aprotic solvent.
C183 the process of C182, wherein the solvent is selected from the group consisting of: dimethyl sulfoxide (DMSO), sulfolane (sulfolane), Dimethylformamide (DMF), and Hexamethylphosphoramide (HMPA).
C184. The method of C183, wherein the solvent is dimethyl sulfoxide (DMSO).
C185. The process according to any one of C181 to C184 wherein the polysaccharide is reacted with 0.01 to 10.0 molar equivalents of an oxidizing agent.
C186. The method of any one of C181 to C185, wherein the oxidizing agent is a periodate salt.
C187. The process of C186, wherein the periodate is sodium periodate.
C188. The method of any one of C181 to C187, wherein the oxidation reaction of step (a) is for 1 to 50 hours.
C189. The method of any one of C181 to C188, wherein the temperature of the oxidation reaction is maintained at about 2 ℃ to about 25 ℃.
C190. The method of any one of C181 to C189, wherein the oxidation reaction is performed in a buffer selected from the group consisting of: sodium phosphate, potassium phosphate, 2- (N-morpholino) ethanesulfonic acid (MES) and Bis-Tris.
C191. The method of claim C190, wherein the buffer is at a concentration of about 1mM to about 500 mM.
C192. The method of any one of C181 to C191, wherein the oxidation reaction is carried out at a pH of about 4.0 to about 8.0.
C193. The method of C181, wherein the oxidizing agent is 2,2,6, 6-tetramethyl-1-piperidinyloxy (TEMPO).
C194. The process of C193 wherein N-chlorosuccinimide (NCS) is a co-oxidant.
C195. The method of any one of C181 to C194, wherein step (a) further comprises quenching the oxidation reaction by adding a quencher.
C196. The method of any one of C182 to C195, wherein the concentration of the polysaccharide is about 0.1mg/mL to about 10.0 mg/mL.
C197. The method of any one of C181 to C196, wherein the activated polysaccharide has an oxidation degree of 5 to 25.
C198. The method of any one of C181 to C197, wherein the method further comprises the step of freeze-drying the activated polysaccharide.
C199. The method of C188, wherein the activated polysaccharide is freeze-dried in the presence of a sugar selected from the group consisting of: sucrose, trehalose, raffinose, stachyose, melezitose, dextran, mannitol, lactitol, and xylitol.
C200. The method of any one of C181 to C199, wherein step (b) comprises:
(1) mixing the activated polysaccharide with a carrier protein, and
(2) reacting the mixed activated polysaccharide and carrier protein with a reducing agent to form a GBS capsular polysaccharide-carrier protein conjugate.
C201. The method of C200, wherein the activated polysaccharide in step (2) is at a concentration of about 0.1mg/mL to about 10.0 mg/mL.
C202. The method of C200 or C201, wherein the initial ratio (weight/weight) of the activated polysaccharide to carrier protein is about 5: 1 to 0.1: 1.
C203. the method of any one of C200 to C202, wherein the reducing agent is selected from the group consisting of: sodium cyanoborohydride, sodium triacetoxyborohydride, sodium borohydride and zinc borohydride in the presence of Bronsted acid or Lewis acid, pyridine borane, 2-picoline borane, 2,6-diborane-methanol, dimethylamine-borane, t-BuMeiPrN-BH3benzylamine-BH3Or 5-ethyl-2-methylpyridine borane (PEMB).
C204. The process of C203, wherein the reducing agent is sodium cyanoborohydride.
C205. The method of any one of C200 to C204, wherein the amount of reducing agent is about 0.1 to about 10.0 molar equivalents.
C206. The method of any one of C200 to C205, wherein the duration of the reduction reaction of step (2) is 1 hour to 60 hours.
C207. The method according to any one of C200 to C206, wherein the temperature of the reduction reaction is maintained at 10 ℃ to 40 ℃.
C208. The method according to any one of C181 to C207, wherein the method further comprises a step of capping the unreacted aldehyde by adding borohydride (step (C)).
C209. The method of C208, wherein the amount of borohydride is about 0.1 to about 10.0 molar equivalents.
C210. The method of C208, wherein the borohydride is selected from the group consisting of: sodium borohydride (NaBH)4) Sodium cyanoborohydride, lithium borohydride, potassium borohydride, tetrabutylammonium borohydride (tetrabutylammonium borohydride), calcium borohydride and magnesium borohydride.
C211. The method of C292, wherein the borohydride is sodium borohydride (NaBH)4)。
C212. The method of any one of C207 to C211, wherein the duration of the capping step is 0.1 to 10 hours.
C213. The method of any one of C207 to C212, wherein the temperature of the capping step is maintained at about 15 ℃ to about 45 ℃.
C214. The method of any one of C181 to C213, wherein the method further comprises a step of purifying the polysaccharide-protein conjugate.
C215. The method of any one of C181 to C214, wherein the polysaccharide-protein conjugate comprises less than about 40% free polysaccharide compared to the total amount of polysaccharide.
C216. The method of any one of C181 to C215, wherein the ratio (weight/weight) of polysaccharide to carrier protein in the conjugate is about 0.5 to about 3.0.
C217. The method of any one of C181 to C216, wherein the conjugate has a degree of conjugation of 2 to 15.
C218. A method of making a polysaccharide-protein conjugate comprising the steps of:
(a) reacting the isolated GBS capsular polysaccharide with an oxidizing agent;
(b) quenching the oxidation reaction of step (a) by adding a quenching agent to produce an activated GBS capsular polysaccharide;
(c) mixing the activated GBS capsular polysaccharide with a carrier protein,
(d) reacting the mixed activated GBS capsular polysaccharide and carrier protein with a reducing agent to form a GBS capsular polysaccharide-carrier protein conjugate, and
(e) by adding sodium borohydride (NaBH)4) Capping the unreacted aldehyde, wherein steps (c) and (d) are performed in DMSO.
The invention also relates to the following embodiments:
1. an immunogenic polysaccharide-protein conjugate comprising a Group B Streptococcus (GBS) capsular polysaccharide and a carrier protein, wherein the capsular polysaccharide has a sialic acid level of greater than about 60%, greater than about 95%, or about 100%.
2. The immunogenic conjugate of embodiment 1, wherein the capsular polysaccharide is selected from the group consisting of: serotypes Ia, Ib, II, III, IV, V, VI, VII, VIII and IX.
3. The immunogenic conjugate of embodiment 1 or 2, wherein the capsular polysaccharide has at least about 0.6, 0.65, 0.7, 0.75, 0.8, 0.85, 0.9 or 0.95mM sialic acid per mM polysaccharide.
4. The immunogenic conjugate of any one of embodiments 1-3, wherein the capsular polysaccharide has a molecular weight of about 5kDa to about 1,000kDa, about 25kDa to about 750kDa, about 25kDa to about 400kDa, about 25kDa to about 200kDa, or about 100kDa to about 400 kDa.
5. The immunogenic conjugate of any one of embodiments 1-4, wherein the conjugate has a molecular weight of about 300kDa to about 20,000kDa, about 1,000kDa to about 15,000kDa, or about 1,000kDa to about 10,000 kDa.
6. The immunogenic conjugate of any one of embodiments 1-5, wherein the capsular polysaccharide is about 0% to about 40% O-acetylated.
7. The immunogenic conjugate of any one of embodiments 1-6, wherein the capsular polysaccharide is less than about 5%, less than about 4%, less than about 3%, less than about 2%, or less than about 1% O-acetylated.
8. The immunogenic conjugate of any one of embodiments 1-7, wherein the capsular polysaccharide has at least about 0.1, 0.2, 0.3, 0.35, or about 0.4mM O-acetate per mM saccharide repeat unit.
9. The immunogenic conjugate of any one of embodiments 1-7, wherein the capsular polysaccharide has at least about 0.01, 0.02, 0.03, 0.04, or 0.05mM O-acetate per mM saccharide repeat unit.
10. The immunogenic conjugate of any one of embodiments 1-9, wherein the carrier protein is CRM197Or tetanus toxoid.
11. The immunogenic conjugate of embodiment 10, wherein the carrier protein is CRM197
12. A process for isolating capsular polysaccharides comprising reacting an organic reagent with a cell broth comprising a capsular polysaccharide producing bacterium.
13. The method of embodiment 12, wherein the bacterium is not lysed.
14. The method of embodiment 12 or 13, wherein the bacteria are heat killed.
15. The method of any one of embodiments 12-14, wherein the method further comprises a centrifugation step to provide a cell paste.
16. The method of any one of embodiments 12-15, wherein the method further comprises a filtration step.
17. The method of embodiment 16, wherein the filtration step is diafiltration.
18. The method of any one of embodiments 12-17, wherein the capsular polysaccharide producing bacterium is selected from the group consisting of: streptococcus agalactiae (Streptococcus agalactiae), Streptococcus pneumoniae (Streptococcus pneumoniae), Staphylococcus aureus (Staphylococcus aureus), Neisseria meningitidis (Neisseria meningitidis), Escherichia coli (Escherichia coli), Salmonella typhi (Salmonella typhi), Haemophilus influenzae (Haemophilus influenzae), Klebsiella pneumoniae (Klebsiella pneumoniae), Enterococcus faecium (Enterococcus faecalis), and Enterococcus faecalis (Enterococcus faecalis).
19. The method of embodiment 18, wherein the bacterium is streptococcus agalactiae.
20. The method of any of embodiments 12-19, wherein the organic reagent is a derivatized hydroxylamine compound.
21. The method of any one of embodiments 12-20, wherein the hydroxylamine is any of the hydroxylamines listed in table 2 of example 2.
22. The method of any one of embodiments 12-21, wherein hydroxylamine is selected from the group consisting of: dibenzylhydroxylamine; diethylhydroxylamine; a hydroxylamine; ethylene diamine; triethylenetetramine; 1,1,4,7,10,10 hexamethyl triethylenetetramine; and 2,6,10 trimethyl 2,6,10 triazaundecane.
23. The method of any one of embodiments 12-22, wherein the concentration of hydroxylamine is from about 5mM to about 200 mM.
24. The method of any one of embodiments 12-23, wherein the pH of the reaction is about 5.5 to about 9.5.
25. The method of any one of embodiments 12-24, wherein the reacting is carried out at a temperature of about 20 ℃ to about 85 ℃.
26. The method of any one of embodiments 12-26, wherein the reaction time is from about 10 hours to about 90 hours.
27. A process for making an immunogenic polysaccharide-protein conjugate according to any one of embodiments 1 to 11, wherein the capsular polysaccharide is isolated according to the process of any one of embodiments 12 to 26.
28. An immunogenic polysaccharide-protein conjugate comprising a capsular polysaccharide manufactured by the process of any one of embodiments 12 to 26.
29. An immunogenic composition comprising an immunogenic polysaccharide-protein conjugate according to any one of embodiments 1 to 11 or 28.
30. An immunogenic composition comprising a polysaccharide-protein conjugate, wherein the conjugate comprises a capsular polysaccharide from Group B Streptococcus (GBS) serotype IV and at least one additional serotype selected from the group consisting of: ia. Ib, II, III, V, VI, VII, VIII and IX.
31. An immunogenic composition comprising a polysaccharide-protein conjugate, wherein the conjugate comprises capsular polysaccharides from GBS serotypes Ia, Ib, II, III and IV.
32. An immunogenic composition comprising a polysaccharide-protein conjugate, wherein the conjugate comprises capsular polysaccharides from GBS serotypes Ia, Ib, II, III and V.
33. An immunogenic composition comprising a polysaccharide-protein conjugate, wherein the conjugate comprises capsular polysaccharides from GBS serotypes Ia, Ib, II, III, IV and V.
34. An immunogenic composition comprising a polysaccharide-protein conjugate comprising at least four GBS capsular polysaccharide serotypes selected from the group consisting of: ia. Ib, II, III, IV, V, VI, VII, VIII and IX.
35. The immunogenic composition of embodiment 34, wherein the composition comprises GBS capsular polysaccharide serotype V.
36. The immunogenic composition of embodiment 34 or 35, wherein the composition does not have immune interference.
37. The immunogenic composition of any one of embodiments 29-36, wherein the composition further comprises a pharmaceutically acceptable excipient, buffer, stabilizer, adjuvant, cryoprotectant, salt, divalent cation, non-ionic detergent, free radical oxidation inhibitor, carrier, or mixture thereof.
38. The immunogenic composition of any one of embodiments 29-37, wherein the composition further comprises a buffering agent.
39. The immunogenic composition of embodiment 38, wherein the buffer is selected from the group consisting of: HEPES, PIPES, MES, Tris (trometamol), phosphate, acetate, borate, citrate, glycine, histidine and succinate.
40. The immunogenic composition of embodiment 39, wherein the buffering agent is histidine.
41. The immunogenic composition of any one of embodiments 29-40, wherein the composition further comprises a surfactant.
42. The immunogenic composition of embodiment 41, wherein the surfactant is selected from the group consisting of: polyoxyethylene sorbitan fatty acid ester, polysorbate-80, polysorbate-60, polysorbate-40, polysorbate-20 and polyoxyethylene alkyl ether.
43. The immunogenic composition of embodiment 42, wherein the surfactant is polysorbate-80.
44. The immunogenic composition of any one of embodiments 29-43, wherein the composition further comprises an excipient.
45. The immunogenic composition of embodiment 44, wherein the excipient is selected from the group consisting of: starch, glucose, lactose, sucrose, trehalose, raffinose, stachyose, melezitose, dextran, mannitol, lactitol, arabitol, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, glycine, arginine, lysine, sodium chloride (NaCl), dried skim milk, glycerol, propylene glycol, water and ethanol.
46. The immunogenic composition of embodiment 45, wherein the excipient is sodium chloride.
47. The immunogenic composition of any one of embodiments 29-46, wherein the composition further comprises an adjuvant.
48. The immunogenic composition of embodiment 47, wherein the adjuvant is an aluminum-based adjuvant or QS-21.
49. The immunogenic composition of embodiment 48, wherein the aluminum-based adjuvant is selected from the group consisting of: aluminum phosphate, aluminum hydroxyphosphate, and aluminum hydroxide.
50. The immunogenic composition of embodiment 49, wherein the adjuvant is aluminum phosphate.
51. The immunogenic composition of embodiment 49, wherein the adjuvant is aluminum hydroxyphosphate.
52. The immunogenic composition of any one of embodiments 29-51, wherein the composition comprises a buffer, a surfactant, an excipient, and optionally an adjuvant, wherein the composition is buffered to a pH of about 6.0 to about 7.0.
53. The immunogenic composition of any one of embodiments 29-52, wherein the composition comprises histidine, polysorbate-80, sodium chloride, and optionally aluminum phosphate, wherein the composition is buffered to a pH of about 6.0 to about 7.0.
54. The immunogenic composition of any one of embodiments 29-53, wherein the composition comprises about 10mM to about 25mM histidine, about 0.01% to about 0.03% (v/w) polysorbate-80, about 10mM to about 250mM sodium chloride, and optionally about 0.25mg/ml to about 0.75mg/ml aluminum as aluminum phosphate.
55. The immunogenic composition of any one of embodiments 29-54, wherein the composition comprises a dose of about 5mcg/ml to about 50 mcg/ml.
56. The immunogenic composition of any one of embodiments 29-55, wherein the composition is lyophilized, optionally in the presence of at least one excipient.
57. The immunogenic composition of embodiment 56, wherein the at least one excipient is selected from the group consisting of: starch, glucose, lactose, sucrose, trehalose, raffinose, stachyose, melezitose, dextran, mannitol, lactitol, arabitol, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, glycine, arginine, lysine, sodium chloride (NaCl), dried skim milk, glycerol, propylene glycol, water and ethanol.
58. The immunogenic composition of embodiment 57, wherein the at least one excipient is sucrose.
59. The immunogenic composition of any one of embodiments 56-58, wherein the composition comprises about 1% (w/v) to about 10% (w/v) of the at least one excipient.
60. The immunogenic composition of any one of embodiments 56-59, wherein the composition comprises additional excipients.
61. The immunogenic composition of embodiment 60, wherein the additional excipient is mannitol, or glycine.
62. The immunogenic composition of embodiment 60 or 61, wherein the composition comprises about 1% (w/v) to about 10% (w/v) additional excipient.
63. The immunogenic composition of any one of embodiments 29-66, wherein the composition is reconstituted with water, water for injection (WFI), an adjuvant suspension, or saline.
64. The immunogenic composition of any one of embodiments 29-63 for use as a medicament.
65. The immunogenic composition of any one of embodiments 29-63, for use in a method of eliciting an immune response to GBS in a subject.
66. The immunogenic composition of embodiment 65, wherein the subject is a woman scheduled to become pregnant or a pregnant woman.
67. The immunogenic composition of embodiment 66, wherein the female is in the second half of her pregnancy, at least at week 20 of pregnancy, or between weeks 27 and 36 of pregnancy.
68. The immunogenic composition of embodiment 65, wherein the subject is an adult of 50 years or older, an adult of 65 years or older, or an adult of 85 years or older.
69. The immunogenic composition of any one of embodiments 65-68, wherein the subject is immunocompromised.
70. The immunogenic composition of embodiment 69, wherein the subject has a medical condition selected from the group consisting of: obesity, diabetes, HIV infection, cancer, cardiovascular disease, or liver disease.
71. The immunogenic composition of any one of embodiments 65-70, wherein the group B streptococcus is Streptococcus agalactiae.
72. A method of eliciting an immune response against group B streptococcus comprising administering to a subject an effective amount of an immunogenic composition according to any one of embodiments 29 to 63.
73. A method of preventing or alleviating a group B streptococcus-associated disease or condition in a subject, comprising administering to the subject an effective amount of an immunogenic composition according to any one of embodiments 29 to 63.
74. The method of embodiment 72 or 73, wherein the subject is a female intended to be pregnant or a pregnant female.
75. The method of embodiment 74, wherein the female is in the second half of her pregnancy, at least at week 20 of pregnancy, or between weeks 27 and 36 of pregnancy.
76. The method of embodiment 72 or 73, wherein the subject is an adult of 50 years or older, an adult of 65 years or older, or an adult of 85 years or older.
77. The method of any one of embodiments 72-76, wherein the subject is immunocompromised.
78. The method of embodiment 77, wherein the subject has a medical condition selected from the group consisting of: obesity, diabetes, HIV infection, cancer, cardiovascular disease, or liver disease.
79. The method of any one of embodiments 72-78, wherein the group B streptococcus is Streptococcus agalactiae.
80. An antibody that binds to a capsular polysaccharide in an immunogenic conjugate according to any one of embodiments 1 to 11 or 28.
81. A composition comprising the antibody of embodiment 80.
82. A method of producing an antibody comprising administering to a subject an immunogenic composition as in any one of embodiments 29 to 63.
83. An antibody produced by the method of embodiment 82.
84. A method of conferring passive immunity to a subject, comprising the steps of:
(a) generating an antibody preparation using the immunogenic composition of any one of embodiments 29 to 63, and
(b) the antibody formulation is administered to a subject to confer passive immunity.
85. A method of making an immunogenic polysaccharide-protein conjugate according to any one of embodiments 1 to 11 or 28 comprising the steps of:
(a) Reacting a GBS capsular polysaccharide with an oxidizing agent to produce an activated polysaccharide; and
(b) reacting the activated polysaccharide with a carrier protein to produce a polysaccharide-protein conjugate.
86. The method of embodiment 85, wherein step (b) is performed in a polar aprotic solvent.
87. The method of embodiment 86, wherein the solvent is selected from the group consisting of: dimethyl sulfoxide (DMSO), sulfolane, Dimethylformamide (DMF), and Hexamethylphosphoramide (HMPA).
88. The method of embodiment 87, wherein the solvent is dimethyl sulfoxide (DMSO).
89. The method of any one of embodiments 85-88, wherein the polysaccharide is reacted with 0.01 to 10.0 molar equivalents of an oxidizing agent.
90. The method of any one of embodiments 85-89, wherein the oxidizing agent is a periodate salt.
91. The method of embodiment 90, wherein the periodate is sodium periodate.
92. The method of any one of embodiments 85 to 91, wherein the oxidation reaction of step (a) is from 1 hour to 50 hours.
93. The method of any one of embodiments 85 to 92, wherein the temperature of the oxidation reaction is maintained at about 2 ℃ to about 25 ℃.
94. The method of any one of embodiments 85-93, wherein the oxidation reaction is performed in a buffer selected from the group consisting of: sodium phosphate, potassium phosphate, 2- (N-morpholino) ethanesulfonic acid (MES) and Bis-Tris.
95. The method of embodiment 94, wherein the buffer is at a concentration of about 1mM to about 500 mM.
96. The method of any one of embodiments 85 to 95, wherein the oxidation reaction is performed at a pH of about 4.0 to about 8.0.
97. The method of embodiment 85 wherein the oxidizing agent is 2,2,6, 6-tetramethyl-1-piperidinyloxy (TEMPO).
98. The method of embodiment 97, wherein N-chlorosuccinimide (NCS) is a co-oxidant.
99. The method of any one of embodiments 85 to 98, wherein step (a) further comprises quenching the oxidation reaction by adding a quencher.
100. The method of any one of embodiments 85 to 99, wherein the concentration of the polysaccharide is about 0.1mg/mL to about 10.0 mg/mL.
101. The method of any one of embodiments 85 to 100, wherein the activated polysaccharide has an oxidation degree of 5 to 25.
102. The method of any one of embodiments 85 to 101, wherein the method further comprises the step of freeze drying the activated polysaccharide.
103. The method of embodiment 102, wherein the activated polysaccharide is freeze-dried in the presence of a sugar selected from the group consisting of: sucrose, trehalose, raffinose, stachyose, melezitose, dextran, mannitol, lactitol, and xylitol.
104. The method of any one of embodiments 85-103, wherein step (b) comprises:
(1) mixing the activated polysaccharide with a carrier protein, and
(2) reacting the mixed activated polysaccharide and carrier protein with a reducing agent to form a GBS capsular polysaccharide-carrier protein conjugate.
105. The method of embodiment 104, wherein the activated polysaccharide in step (2) is at a concentration of about 0.1mg/mL to about 10.0 mg/mL.
106. The method of embodiment 104 or 105, wherein the initial ratio (weight/weight) of the activated polysaccharide to carrier protein is about 5:1 to 0.1: 1.
107. The method as recited in any one of embodiments 104-106, wherein the reducing agent is selected from the group consisting of: sodium cyanoborohydride, sodium triacetoxyborohydride, sodium borohydride in the presence of Bronsted acid or Lewis acid, zinc borohydride, pyridine borane, 2-picoline borane, 2, 6-diborane-methanol, dimethylamine-borane, t-BuMeiPrN-BH3benzylamine-BH3Or 5-ethyl-2-picoline borane (PEMB).
108. The method of embodiment 107 wherein the reducing agent is sodium cyanoborohydride.
109. The method of any one of embodiments 104-108, wherein the amount of reducing agent is about 0.1 to about 10.0 molar equivalents.
110. The method as in any one of embodiments 104-109, wherein the duration of the reduction reaction of step (2) is from 1 hour to 60 hours.
111. The method as in any one of embodiments 104-110, wherein the temperature of the reduction reaction is maintained between 10 ℃ and 40 ℃.
112. The method as in any one of embodiments 104-111, wherein the method further comprises a step of capping unreacted aldehyde by adding borohydride (step (c)).
113. The method of embodiment 112, wherein the amount of borohydride is about 0.1 to about 10.0 molar equivalents.
114. The method of embodiment 112, wherein the borohydride is selected from the group consisting of: sodium borohydride (NaBH)4) Sodium cyanoborohydride, lithium borohydride, potassium borohydride, tetrabutylammonium borohydride, calcium borohydride and magnesium borohydride.
115. The method of embodiment 114, wherein the borohydride is sodium borohydride (NaBH)4)。
116. The method as in any one of embodiments 112-114, wherein the capping step has a duration of 0.1 hour to 10 hours.
117. The method of any one of embodiments 112-116, wherein the temperature of the capping step is maintained at about 15 ℃ to about 45 ℃.
118. The method of any one of embodiments 85-117, wherein the method further comprises a step of purifying the polysaccharide-protein conjugate.
119. The method of any one of embodiments 85-118, wherein the polysaccharide-protein conjugate comprises less than about 40% free polysaccharide compared to the total amount of polysaccharide.
120. The method of any one of embodiments 85-119, wherein the ratio (weight/weight) of polysaccharide to carrier protein in the conjugate is about 0.5 to about 3.0.
121. The method of any one of embodiments 85 to 120, wherein the conjugate has a degree of conjugation of 2 to 15.
122. A method of making a polysaccharide-protein conjugate comprising the steps of:
(a) reacting the isolated GBS capsular polysaccharide with an oxidizing agent;
(b) quenching the oxidation reaction of step (a) by adding a quenching agent to produce an activated GBS capsular polysaccharide;
(c) mixing the activated GBS capsular polysaccharide with a carrier protein,
(d) reacting the mixed activated GBS capsular polysaccharide and carrier protein with a reducing agent to form a GBS capsular polysaccharide-carrier protein conjugate, and
(e) by adding sodium borohydride (NaBH)4) To end-cap the unreacted aldehyde(s),
wherein steps (c) and (d) are performed in DMSO.

Claims (16)

1. An immunogenic polysaccharide-protein conjugate comprising a Group B Streptococcus (GBS) capsular polysaccharide and a carrier protein, wherein the capsular polysaccharide has a sialic acid level of greater than about 60%, greater than about 95%, or about 100%.
2. The immunogenic conjugate of claim 1, wherein the capsular polysaccharide is selected from the group consisting of: serotypes Ia, Ib, II, III, IV, V, VI, VII, VIII and IX.
3. A process for isolating capsular polysaccharides comprising reacting an organic reagent with a cell broth comprising a capsular polysaccharide producing bacterium.
4. A process for making the immunogenic polysaccharide-protein conjugate of claim 1 or 2, wherein the capsular polysaccharide is isolated according to the process of claim 3.
5. An immunogenic polysaccharide-protein conjugate comprising a capsular polysaccharide manufactured by the process of claim 3.
6. An immunogenic composition comprising the immunogenic polysaccharide-protein conjugate of any one of claims 1 to 2 or 5.
7. An immunogenic composition comprising a polysaccharide-protein conjugate, wherein the conjugate comprises a capsular polysaccharide from Group B Streptococcus (GBS) serotype IV and at least one additional serotype selected from the group consisting of: ia. Ib, II, III, V, VI, VII, VIII and IX.
8. An immunogenic composition comprising a polysaccharide-protein conjugate, wherein the conjugate comprises capsular polysaccharides from GBS serotypes Ia, Ib, II, III and IV.
9. An immunogenic composition comprising a polysaccharide-protein conjugate comprising at least four GBS capsular polysaccharide serotypes selected from the group consisting of: ia. Ib, II, III, IV, V, VI, VII, VIII and IX.
10. An antibody that binds to the capsular polysaccharide in the immunogenic conjugate of any one of claims 1 to 2 or 5.
11. A composition comprising the antibody of claim 10.
12. A method of producing an antibody comprising administering to a subject the immunogenic composition of any one of claims 6 to 9.
13. An antibody produced by the method of claim 12.
14. Use of an immunogenic composition according to any one of claims 6 to 9 in the manufacture of a medicament for conferring passive immunity to a subject.
15. A method of making the immunogenic polysaccharide-protein conjugate of any one of claims 1 to 2 or 5 comprising the steps of:
(a) reacting a GBS capsular polysaccharide with an oxidizing agent to produce an activated polysaccharide; and
(b) reacting the activated polysaccharide with a carrier protein to produce a polysaccharide-protein conjugate.
16. A method of making a polysaccharide-protein conjugate comprising the steps of:
(a) Reacting the isolated GBS capsular polysaccharide with an oxidizing agent;
(b) quenching the oxidation reaction of step (a) by adding a quenching agent to produce an activated GBS capsular polysaccharide;
(c) mixing the activated GBS capsular polysaccharide with a carrier protein,
(d) reacting the mixed activated GBS capsular polysaccharide and carrier protein with a reducing agent to form a GBS capsular polysaccharide-carrier protein conjugate, and
(e) by adding sodium borohydride (NaBH)4) To end-cap the unreacted aldehyde(s),
wherein steps (c) and (d) are performed in DMSO.
CN202111336096.5A 2015-05-04 2016-04-29 Group B streptococcal polysaccharide-protein conjugates, methods of making conjugates, immunogenic compositions containing conjugates, and uses thereof Pending CN114010775A (en)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US201562156500P 2015-05-04 2015-05-04
US62/156,500 2015-05-04
US201562237813P 2015-10-06 2015-10-06
US201562237820P 2015-10-06 2015-10-06
US62/237,820 2015-10-06
US62/237,813 2015-10-06
US201662319539P 2016-04-07 2016-04-07
US62/319,539 2016-04-07
CN201680025709.4A CN107624070A (en) 2015-05-04 2016-04-29 B races Streptococcus polysaccharide protein conjugate, the method for manufacturing conjugate, immunogenic composition containing conjugate and application thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN201680025709.4A Division CN107624070A (en) 2015-05-04 2016-04-29 B races Streptococcus polysaccharide protein conjugate, the method for manufacturing conjugate, immunogenic composition containing conjugate and application thereof

Publications (1)

Publication Number Publication Date
CN114010775A true CN114010775A (en) 2022-02-08

Family

ID=55967339

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202111336096.5A Pending CN114010775A (en) 2015-05-04 2016-04-29 Group B streptococcal polysaccharide-protein conjugates, methods of making conjugates, immunogenic compositions containing conjugates, and uses thereof
CN201680025709.4A Pending CN107624070A (en) 2015-05-04 2016-04-29 B races Streptococcus polysaccharide protein conjugate, the method for manufacturing conjugate, immunogenic composition containing conjugate and application thereof

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN201680025709.4A Pending CN107624070A (en) 2015-05-04 2016-04-29 B races Streptococcus polysaccharide protein conjugate, the method for manufacturing conjugate, immunogenic composition containing conjugate and application thereof

Country Status (21)

Country Link
US (4) US10226525B2 (en)
EP (1) EP3292146A1 (en)
JP (3) JP6948951B2 (en)
KR (2) KR102143534B1 (en)
CN (2) CN114010775A (en)
AU (2) AU2016258284C1 (en)
BR (1) BR112017022776A2 (en)
CA (1) CA2928602A1 (en)
CO (1) CO2017011245A2 (en)
IL (2) IL255106B2 (en)
MX (1) MX368897B (en)
MY (1) MY182282A (en)
NZ (1) NZ736238A (en)
PE (2) PE20180172A1 (en)
PH (1) PH12017501898A1 (en)
RU (1) RU2692923C2 (en)
SA (1) SA517390274B1 (en)
SG (2) SG10202101887WA (en)
TW (2) TWI718144B (en)
WO (1) WO2016178123A1 (en)
ZA (1) ZA201807203B (en)

Families Citing this family (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SA115360586B1 (en) 2012-03-09 2017-04-12 فايزر انك Neisseria meningitidis compositions and methods thereof
MX369534B (en) 2013-09-08 2019-11-11 Pfizer Neisseria meningitidis compositions and methods thereof.
PL3096783T3 (en) 2014-01-21 2021-12-13 Pfizer Inc. Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
US11160855B2 (en) * 2014-01-21 2021-11-02 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
IL255106B2 (en) 2015-05-04 2023-04-01 Pfizer Group b streptococcus polysaccharide-protein conjugates, methods for producing conjugates, immunogenic compositions comprising conjugates, and uses thereof
US10751402B2 (en) 2016-11-09 2020-08-25 Pfizer Inc. Immunogenic compositions and uses thereof
CN108144052A (en) * 2016-12-02 2018-06-12 武汉博沃生物科技有限公司 Streptococcus pneumoniae polysaccharides-protein conjugate and its preparation method and purposes
US20190343951A1 (en) * 2017-01-13 2019-11-14 National Research Council Of Canada Method of optimizing peptide immuno-epitope by glycosylation, optimized peptide thereof and its use for conjugate vaccines
US11197921B2 (en) 2017-01-31 2021-12-14 Merck Sharp & Dohme Corp. Methods for making polysaccharide-protein conjugates
AU2018225099A1 (en) 2017-02-24 2019-07-25 Merck Sharp & Dohme Llc Enhancing immunogenicity of Streptococcus pneumoniae polysaccharide-protein conjugates
AU2018225083B2 (en) * 2017-02-24 2023-06-01 Merck Sharp & Dohme Llc Pneumococcal conjugate vaccine formulations
CA3066020A1 (en) 2017-06-16 2018-12-20 Glaxosmithkline Biologicals Sa Method of treatment
EP3668541A4 (en) * 2017-08-16 2021-05-26 Merck Sharp & Dohme Corp. Pneumococcal conjugate vaccine formulations
CN116898959A (en) 2017-09-07 2023-10-20 默沙东有限责任公司 Pneumococcal polysaccharide and its use in immunogenic polysaccharide-carrier protein conjugates
CN111065387B (en) 2017-09-07 2023-08-25 默沙东有限责任公司 Pneumococcal polysaccharide and its use in immunogenic polysaccharide-carrier protein conjugates
PE20201338A1 (en) 2017-12-06 2020-11-25 Merck Sharp & Dohme COMPOSITIONS INCLUDING CONJUGATES OF STREPTOCOCCUS PNEUMONIAE WITH PROTEIN AND METHODS OF USE
CN112041435A (en) * 2018-04-30 2020-12-04 默沙东公司 Method for providing a homogeneous solution of lyophilized mutant diphtheria toxin in dimethyl sulfoxide
US11260119B2 (en) 2018-08-24 2022-03-01 Pfizer Inc. Escherichia coli compositions and methods thereof
KR102120830B1 (en) * 2018-09-20 2020-06-09 고려대학교산학협력단 Novel Group B Streptococcus Serotype Strain and Immunogenic Composition Comprising the Same
CN113453708A (en) 2018-12-19 2021-09-28 默沙东公司 Compositions comprising streptococcus pneumoniae polysaccharide-protein conjugates and methods of use thereof
US20220211859A1 (en) * 2019-05-10 2022-07-07 Glaxosmithkline Biologicals Sa Conjugate production
CN112741901A (en) * 2019-10-31 2021-05-04 北京科兴中维生物技术有限公司 Vaccine containing streptococcus pneumoniae capsular polysaccharide type 5 and preparation method thereof
EP4061412A1 (en) 2019-11-22 2022-09-28 GlaxoSmithKline Biologicals S.A. Dosage and administration of a bacterial saccharide glycoconjugate vaccine
BR112022023261A2 (en) 2020-06-12 2022-12-27 Glaxosmithkline Biologicals Sa BACTERIAL IMMUNIZATION USING NANOPARTICLE VACCINE
EP4165065A2 (en) 2020-06-12 2023-04-19 GlaxoSmithKline Biologicals S.A. Gbs ferritin nanoparticles
CN114073761B (en) * 2020-08-11 2023-11-14 山东大学 Oligosaccharide conjugate and application thereof as saccharide vaccine for preventing group V-B streptococcus infection
US11672761B2 (en) 2020-11-16 2023-06-13 Orcosa Inc. Rapidly infusing platform and compositions for therapeutic treatment in humans
CN112939164B (en) * 2021-03-15 2022-05-24 广西大学 Method for disinfecting marine aquaculture water
CA3221075A1 (en) * 2021-05-28 2022-12-01 Pfizer Inc. Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
CN113480622A (en) * 2021-08-05 2021-10-08 江苏坤力生物制药有限责任公司 Method for preparing and purifying recombinant pneumolysin
CN113651730A (en) * 2021-08-20 2021-11-16 宿迁盛瑞新材料有限公司 Preparation method of bis (octadecyl) hydroxylamine
CN115960962A (en) * 2021-10-09 2023-04-14 江苏金斯瑞蓬勃生物科技有限公司 Protective agent for enhancing stability of lentivirus vector and application thereof
CN113621089B (en) * 2021-10-11 2021-12-17 山东洁壹选生物科技有限公司 Preparation method and application of algal polysaccharide extract
WO2023111826A1 (en) 2021-12-14 2023-06-22 Glaxosmithkline Biologicals Sa Bacterial immunization using qbeta hairpin nanoparticle constructs
CN114015622A (en) * 2021-12-14 2022-02-08 塔里木大学 Streptococcus agalactiae culture medium and preparation method thereof

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0502095D0 (en) * 2005-02-01 2005-03-09 Chiron Srl Conjugation of streptococcal capsular saccharides
US20130273091A1 (en) * 2010-09-16 2013-10-17 Francesco Berti Immunogenic compositions
WO2014053612A1 (en) * 2012-10-03 2014-04-10 Novartis Ag Immunogenic composition

Family Cites Families (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5360897A (en) 1981-08-31 1994-11-01 The University Of Rochester Immunogenic conjugates of streptococcus pneumonial capsular polymer and toxin or in toxiad
CA1247080A (en) 1983-03-08 1988-12-20 Commonwealth Serum Laboratories Commission Antigenically active amino acid sequences
SE8405493D0 (en) 1984-11-01 1984-11-01 Bror Morein IMMUNOGENT COMPLEX AND KITCHEN FOR PREPARING IT AND USING IT AS IMMUNOSTIMENTING AGENTS
US5078996A (en) 1985-08-16 1992-01-07 Immunex Corporation Activation of macrophage tumoricidal activity by granulocyte-macrophage colony stimulating factor
US5057540A (en) 1987-05-29 1991-10-15 Cambridge Biotech Corporation Saponin adjuvant
US4912094B1 (en) 1988-06-29 1994-02-15 Ribi Immunochem Research Inc. Modified lipopolysaccharides and process of preparation
HU212924B (en) 1989-05-25 1996-12-30 Chiron Corp Adjuvant formulation comprising a submicron oil droplet emulsion
US5371197A (en) * 1991-09-24 1994-12-06 Merck & Co., Inc. Protein-dimeric polysaccharide conjugate vaccine
IT1253009B (en) 1991-12-31 1995-07-10 Sclavo Ricerca S R L DETOXIFIED IMMUNOGENIC MUTANTS OF COLERIC TOXIN AND TOXIN LT, THEIR PREPARATION AND USE FOR THE PREPARATION OF VACCINES
NZ249704A (en) 1992-02-11 1996-11-26 Jackson H M Found Military Med A two carrier immunogenic construct comprising a 70+ kd molecule conjugated to at least 1 t-dependent antigen, preparation, compositions containing the construct
JP4163251B2 (en) * 1992-09-24 2008-10-08 ブリガーム・アンド・ウーメンズ・ホスピタル・インコーポレーテッド Group B Streptococcus Type II and Type V Polysaccharide-protein conjugate vaccine
ZA937034B (en) * 1992-09-24 1995-06-23 Brigham & Womens Hospital Group B streptococcus type II and type V polysaccharide-protein conjugate vaccines
EP0616034B1 (en) 1993-03-05 2004-10-20 Wyeth Holdings Corporation Plasmid for production of CRM protein and diphtheria toxin
ES2210262T3 (en) 1993-09-22 2004-07-01 Henry M. Jackson Foundation For The Advancement Of Military Medicine PROCEDURE THAT ALLOWS THE ACTIVATION OF A SOLUBLE GLUCIDE WITH THE HELP OF NEW CIANILANT REAGENTS TO PRODUCE IMMUNOGEN STRUCTURES.
US5571515A (en) 1994-04-18 1996-11-05 The Wistar Institute Of Anatomy & Biology Compositions and methods for use of IL-12 as an adjuvant
US6207646B1 (en) 1994-07-15 2001-03-27 University Of Iowa Research Foundation Immunostimulatory nucleic acid molecules
BE1008978A5 (en) 1994-12-27 1996-10-01 Solvay Adjuvants for vaccines.
WO1996029094A1 (en) 1995-03-22 1996-09-26 Andrew Lees Producing immunogenic constructs using soluble carbohydrates activated via organic cyanylating reagents
GB9622159D0 (en) 1996-10-24 1996-12-18 Solvay Sociutu Anonyme Polyanionic polymers as adjuvants for mucosal immunization
GB9622660D0 (en) 1996-10-31 1997-01-08 Biocine Spa Immunogenic detoxified mutant toxin
US6299881B1 (en) 1997-03-24 2001-10-09 Henry M. Jackson Foundation For The Advancement Of Military Medicine Uronium salts for activating hydroxyls, carboxyls, and polysaccharides, and conjugate vaccines, immunogens, and other useful immunological reagents produced using uronium salts
US6113918A (en) 1997-05-08 2000-09-05 Ribi Immunochem Research, Inc. Aminoalkyl glucosamine phosphate compounds and their use as adjuvants and immunoeffectors
US20010051364A1 (en) * 1997-12-23 2001-12-13 Francis Michon Procedures for the extraction and isolation of bacterial capsular polysaccharides for use as vaccines or linked to proteins as conjugate vaccines
ATE468403T2 (en) * 1997-12-23 2010-06-15 Baxter Healthcare Sa METHOD FOR EXTRACTION AND ISOLATION OF BACTERIAL ENVELOPE POLYSACCHARIDES FOR USE AS A VACCINE OR, COUPLED TO PROTEINS, AS A CONJUGATE VACCINE
US6936258B1 (en) 1999-03-19 2005-08-30 Nabi Biopharmaceuticals Staphylococcus antigen and vaccine
US7115730B1 (en) 1999-04-27 2006-10-03 Chiron Srl Immunogenic detoxified mutant E. coli LT-A-toxin
US7384640B1 (en) 1999-09-30 2008-06-10 Wyeth Holdings Corporation Mutant cholera holotoxin as an adjuvant
CA2411575C (en) 2000-06-08 2015-04-07 Walter Schmidt Immunostimulatory oligodeoxynucleotides
AT410635B (en) 2000-10-18 2003-06-25 Cistem Biotechnologies Gmbh VACCINE COMPOSITION
JP2005508143A (en) 2001-06-07 2005-03-31 ワイス・ホールデイングス・コーポレーシヨン Mutated form of cholera holotoxin as an adjuvant
EP1404368B1 (en) 2001-06-07 2009-12-09 Wyeth Holdings Corporation Mutant forms of cholera holotoxin as an adjuvant
TWI313609B (en) 2001-08-21 2009-08-21 Mitsubishi Tanabe Pharma Corp Pharmaceutical composition for inhibiting the metastasis or preventing the recurrence of malignant tumor
WO2004011027A1 (en) * 2002-07-30 2004-02-05 Baxter International Inc. Chimeric multivalent polysaccharide conjugate vaccines
AU2004220590B2 (en) 2003-03-07 2010-02-18 Inhibitex, Inc. Polysaccharide - Staphylococcal surface adhesin carrier protein conjugates for immunization against nosocomial infections
CA2519511A1 (en) 2003-03-17 2004-09-30 Wyeth Holdings Corporation Mutant cholera holotoxin as an adjuvant and an antigen carrier protein
DE602004017864D1 (en) * 2003-06-23 2009-01-02 Baxter Int CARRIER PROTEINS FOR VACCINES
GB0323103D0 (en) 2003-10-02 2003-11-05 Chiron Srl De-acetylated saccharides
GB0411387D0 (en) * 2004-05-21 2004-06-23 Chiron Srl Analysis of saccharide length
GB0502096D0 (en) 2005-02-01 2005-03-09 Chiron Srl Purification of streptococcal capsular polysaccharide
TWI445545B (en) * 2005-04-08 2014-07-21 Wyeth Corp Multivalent pneumococcal polysaccharide-protein conjugate composition
US7955605B2 (en) 2005-04-08 2011-06-07 Wyeth Llc Multivalent pneumococcal polysaccharide-protein conjugate composition
JP5049264B2 (en) 2005-04-08 2012-10-17 ワイス・エルエルシー Separation of contaminants from Streptococcus pneumoniae polysaccharide by pH manipulation
US7709001B2 (en) 2005-04-08 2010-05-04 Wyeth Llc Multivalent pneumococcal polysaccharide-protein conjugate composition
US20070184072A1 (en) 2005-04-08 2007-08-09 Wyeth Multivalent pneumococcal polysaccharide-protein conjugate composition
GB0522303D0 (en) 2005-11-01 2005-12-07 Chiron Srl Culture method
US20090010959A1 (en) 2005-12-22 2009-01-08 Ralph Leon Biemans Pneumococcal Polysaccharide Conjugate Vaccine
GB0605757D0 (en) 2006-03-22 2006-05-03 Chiron Srl Separation of conjugated and unconjugated components
CN102648979B (en) 2006-10-10 2014-05-14 惠氏公司 Purification of streptococcus pneumoniae type 3 polysaccharides
KR101500771B1 (en) 2007-03-23 2015-03-18 와이어쓰 엘엘씨 Shortened purification process for the production of capsular streptococcus pneumoniae polysaccharides
BRPI0813307C1 (en) * 2007-06-26 2021-05-25 Glaxosmithkline Biologicals Sa immunogenic composition, vaccine, and, process to manufacture the vaccine
GB0818453D0 (en) 2008-10-08 2008-11-12 Novartis Ag Fermentation processes for cultivating streptococci and purification processes for obtaining cps therefrom
JP5453461B2 (en) 2009-03-05 2014-03-26 インターデイジタル パテント ホールディングス インコーポレイテッド Methods and equipment for H (e) NB integrity verification and validation
KR20140119165A (en) 2009-06-22 2014-10-08 와이어쓰 엘엘씨 Compositions and methods for preparing staphylococcus aureus serotype 5 and 8 capsular polysaccharide conjugate immunogenic compositions
WO2011080570A2 (en) 2009-12-29 2011-07-07 Micro Labs Limited Extended release pharmaceutical composition comprising linezolid and process for preparing the same
KR20130098308A (en) 2010-08-23 2013-09-04 셀렉타 바이오사이언시즈, 인크. Targeted multi-epitope dosage forms for induction of an immune response to antigens
CA2752980C (en) 2010-09-23 2018-06-19 Anhydrovac Inc. Non-aqueous synthesis of polysaccharide-protein conjugates for vaccines
WO2012061400A2 (en) 2010-11-01 2012-05-10 University Of Maryland, Baltimore Method for reduction of free polysaccharide in polysaccharide-protein vaccines reactions using ion-exchange matrices
GB201121301D0 (en) * 2011-12-12 2012-01-25 Novartis Ag Method
CN104519910B (en) 2012-03-07 2017-05-03 诺华股份有限公司 Adjuvanted formulations of streptococcus pneumoniae antigens
KR102057217B1 (en) 2012-06-20 2020-01-22 에스케이바이오사이언스 주식회사 Multivalent pneumococcal polysaccharide-protein conjugate composition
CA2881420C (en) * 2012-08-16 2016-11-15 Pfizer Inc. Glycoconjugation processes and compositions
RU2606022C2 (en) 2012-09-07 2017-01-10 Ск Кемикалс Ко., Лтд. Method of producing capsular polysaccharide with pneumococcal serotype
BR112015007126A2 (en) 2012-10-02 2017-08-08 Glaxosmithkline Biologicals Sa composition, method for inducing an immune response, and use of a composition
KR20140075196A (en) 2012-12-11 2014-06-19 에스케이케미칼주식회사 Multivalent pneumococcal polysaccharide-protein conjugate composition
KR20140075201A (en) 2012-12-11 2014-06-19 에스케이케미칼주식회사 Multivalent pneumococcal polysaccharide-protein conjugate composition
WO2014097099A2 (en) * 2012-12-20 2014-06-26 Pfizer Inc. Glycoconjugation process
US10544412B2 (en) * 2013-12-02 2020-01-28 Brandeis University Directed evolution of multivalent glycopeptides that tightly bind to target proteins
KR102049825B1 (en) * 2014-01-21 2019-12-03 화이자 인코포레이티드 Streptococcus pneumoniae capsular polysaccharides and conjugates thereof
SG11201604728XA (en) * 2014-01-21 2016-08-30 Pfizer Immunogenic compositions comprising conjugated capsular saccharide antigens and uses thereof
WO2015121783A1 (en) * 2014-02-14 2015-08-20 Pfizer Inc. Immunogenic glycoprotein conjugates
EP3140414B1 (en) * 2014-05-07 2022-10-26 GlaxoSmithKline Biologicals S.A. Purification of secreted polysaccharides from s. agalactiae
TWI583284B (en) * 2015-02-06 2017-05-11 鴻海精密工業股份有限公司 Protective shell
IL255106B2 (en) * 2015-05-04 2023-04-01 Pfizer Group b streptococcus polysaccharide-protein conjugates, methods for producing conjugates, immunogenic compositions comprising conjugates, and uses thereof
CA2988366C (en) 2015-06-08 2021-12-07 Serum Institute Of India Private Ltd. Methods for improving the adsorption of polysaccharide-protein conjugates and multivalent vaccine formulation obtained thereof.
IL265623B2 (en) 2016-09-30 2023-04-01 Biological E Ltd Multivalent pneumococcal vaccine compositions comprising polysaccharide-protein conjugates
US10751402B2 (en) * 2016-11-09 2020-08-25 Pfizer Inc. Immunogenic compositions and uses thereof
MD20190088A2 (en) * 2017-05-05 2020-04-30 Serum Institute Of India Private Limited Method for removal of impurities from bacterial capsular polysaccharide based preparations

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB0502095D0 (en) * 2005-02-01 2005-03-09 Chiron Srl Conjugation of streptococcal capsular saccharides
US20130273091A1 (en) * 2010-09-16 2013-10-17 Francesco Berti Immunogenic compositions
WO2014053612A1 (en) * 2012-10-03 2014-04-10 Novartis Ag Immunogenic composition

Also Published As

Publication number Publication date
KR102045663B1 (en) 2019-11-15
JP6948951B2 (en) 2021-10-13
US20190142922A1 (en) 2019-05-16
TWI807252B (en) 2023-07-01
JP2018514571A (en) 2018-06-07
AU2016258284C1 (en) 2020-09-03
CN107624070A (en) 2018-01-23
TWI718144B (en) 2021-02-11
KR20190128264A (en) 2019-11-15
US20230346903A1 (en) 2023-11-02
PE20180172A1 (en) 2018-01-22
US20160324950A1 (en) 2016-11-10
US10946086B2 (en) 2021-03-16
AU2016258284B2 (en) 2019-01-17
IL255106B2 (en) 2023-04-01
US20210145957A1 (en) 2021-05-20
CA2928602A1 (en) 2016-11-04
US10226525B2 (en) 2019-03-12
CO2017011245A2 (en) 2018-03-28
IL255106B (en) 2022-12-01
KR20170138568A (en) 2017-12-15
TW201704254A (en) 2017-02-01
RU2692923C2 (en) 2019-06-28
IL297740A (en) 2022-12-01
EP3292146A1 (en) 2018-03-14
AU2016258284A1 (en) 2017-11-02
KR102143534B1 (en) 2020-08-12
AU2019202680A1 (en) 2019-05-09
NZ736238A (en) 2022-07-01
PE20220774A1 (en) 2022-05-16
SG11201708242YA (en) 2017-11-29
RU2017136859A3 (en) 2019-06-04
BR112017022776A2 (en) 2018-07-10
AU2019202680B2 (en) 2020-05-14
SG10202101887WA (en) 2021-03-30
MX368897B (en) 2019-10-21
TW202135856A (en) 2021-10-01
RU2017136859A (en) 2019-06-04
IL255106A0 (en) 2017-12-31
MX2017014127A (en) 2018-03-01
JP2024009978A (en) 2024-01-23
WO2016178123A1 (en) 2016-11-10
JP2022008445A (en) 2022-01-13
ZA201807203B (en) 2022-05-25
SA517390274B1 (en) 2022-12-22
PH12017501898A1 (en) 2018-03-05
MY182282A (en) 2021-01-18

Similar Documents

Publication Publication Date Title
US20230346903A1 (en) Group b streptococcus polysaccharide-protein conjugates, methods for producing conjugates, immunogenic compositions comprising conjugates, and uses thereof
JP7295206B2 (en) Immunogenic compositions and uses thereof
US20230321212A1 (en) Group b streptococcus polysaccharide-protein conjugates, methods for producing conjugates, immunogenic compositions comprising conjugates, and uses thereof
CN116744964A (en) Group B streptococcus polysaccharide-protein conjugates, methods for producing conjugates, immunogenic compositions comprising conjugates, and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40065014

Country of ref document: HK